EP1136503A1 - Neuartige, physiologisch wirksame substanz, verfahren zu deren herstellung und anwendung derselben - Google Patents

Neuartige, physiologisch wirksame substanz, verfahren zu deren herstellung und anwendung derselben Download PDF

Info

Publication number
EP1136503A1
EP1136503A1 EP99973037A EP99973037A EP1136503A1 EP 1136503 A1 EP1136503 A1 EP 1136503A1 EP 99973037 A EP99973037 A EP 99973037A EP 99973037 A EP99973037 A EP 99973037A EP 1136503 A1 EP1136503 A1 EP 1136503A1
Authority
EP
European Patent Office
Prior art keywords
senr
seq
polypeptide
salt
precursor protein
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Withdrawn
Application number
EP99973037A
Other languages
English (en)
French (fr)
Inventor
Masaaki Mori
Michiko Abe
Yukio Takeda Yakuhin SHIMOMURA
Tsukasa Sugo
Chieko Kitada
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Takeda Pharmaceutical Co Ltd
Original Assignee
Takeda Chemical Industries Ltd
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Takeda Chemical Industries Ltd filed Critical Takeda Chemical Industries Ltd
Publication of EP1136503A1 publication Critical patent/EP1136503A1/de
Withdrawn legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/575Hormones
    • C07K14/57509Corticotropin releasing factor [CRF] (Urotensin)
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2333/00Assays involving biological materials from specific organisms or of a specific nature
    • G01N2333/435Assays involving biological materials from specific organisms or of a specific nature from animals; from humans
    • G01N2333/705Assays involving receptors, cell surface antigens or cell surface determinants
    • G01N2333/72Assays involving receptors, cell surface antigens or cell surface determinants for hormones
    • G01N2333/726G protein coupled receptor, e.g. TSHR-thyrotropin-receptor, LH/hCG receptor, FSH
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2500/00Screening for compounds of potential therapeutic value

Definitions

  • the present invention relates to a novel polypeptide having a ligand activity to an SENR (sensory epithelium neuropeptide-like receptor), which is a G protein-coupled receptor protein, a DNA encoding the same, etc.
  • SENR sensor epithelium neuropeptide-like receptor
  • G proteins guanine nucleotide-binding proteins
  • hormones, neurotransmitters and the like interact with G protein-coupled receptor protein and, via the interactive reaction, regulate important functions for a living body such as maintenance of homeostasis, reproduction, development of individuals, metabolism, growth, control of the nervous, circulatory, immune, digestive or metabolic system, sensory adaptation, or the like.
  • receptor proteins to various hormones or neurotransmitters for regulating the functions in vivo and these receptor proteins play pivotal roles for regulating these functions.
  • receptor proteins to various hormones or neurotransmitters for regulating the functions in vivo and these receptor proteins play pivotal roles for regulating these functions.
  • PCR polymerase chain reaction
  • G protein-coupled receptor proteins have been found one after another by random sequencing of genomic DNA or cDNA (Nomura, N., et al., DNA Research, 1, 27-35, 1994). Only general method for determining ligands to these G protein-coupled receptor proteins was to deduce ligands from similarity in primary structure of G protein-coupled receptor proteins. However, many orphan G protein-coupled receptor proteins have low homology to known receptors.
  • the structure of a ligand is determined by selecting an agonist to the receptor from compounds artificially synthesized, verifying that the receptor is expressed in the receptor cDNA-transduced cells using the agonist as a probe, then searching an activated substance of intracellular signal transduction system similar to the agonist and purifying the substance.
  • SENR is reported to be one of the orphan G protein-coupled receptors (Tal, M. et al., Biochem. Biophys. Res. Commun., 209, 752-759, 1995).
  • the SENR has a low homology to somatostatin receptor (SSTR4) but it was unknown what its ligand was.
  • SSTR4 somatostatin receptor
  • GPR14 reported by Marchese, A., et al. (Marchese, A., Genomics, 29, 335-344, 1995) is the same receptor as the SENR.
  • Ligands to the SENR which is a G protein-coupled receptor expressed in the organ such as central nervous system, circulatory system, genital system, immune system, digestive apparatus, urinary system, sensory organs and the like, are considered to be useful as drugs but the structure and functions are unknown yet.
  • the present inventors have succeeded in screening a polypeptide recognized by the receptor protein as a ligand under the index of a specific cell stimulating (signal transduction) activity measured, etc., using a cell in which an SENR-encoding cDNA has been expressed by an appropriate means.
  • the inventors have further found that a compound that can alter the binding property between the activator ligand and an SENR described above can be screened.
  • the present invention relates to the following features.
  • the term "substantially the same” is used to mean that the activities of a polypeptide or protein, e.g., the binding activity between a ligand and a receptor (SENR), physiological properties, etc. are substantially the same.
  • Substitution, deletion, addition or insertion of an amino acid does not often cause any significant change in physiological properties or chemical properties of a polypeptide or protein; in this case, such a polypeptide that undergoes substitution, deletion, addition or insertion is considered to be substantially the same as the polypeptide that does not undergo substitution, deletion, addition or insertion.
  • Substantially the same substituent of an amino acid in the amino acid sequence can be selected from, e.g., other amino acids of the class to which the amino acid belongs.
  • non-polar (hydrophobic) amino acids examples include alanine, leucine, isoleucine, valine, proline, phenylalanine, tryptophan, methionine and the like.
  • polar (neutral) amino acids are glycine, serine, threonine, cysteine, tyrosine, asparagine, glutamine, and the like.
  • positively charged (basic) amino acids examples of positively charged (basic) amino acids are arginine, lysine, histidine, and the like.
  • negatively charged (acidic) amino acids include aspartic acid, glutamic acid, and the like.
  • polypeptide of the present invention is a ligand to SENR and refers specifically to the polypeptide containing the same or substantially the same amino acid sequence represented by SEQ ID NO:7, its amide or ester, or a salt thereof, etc.
  • polypeptide of the present invention its amide or ester, or a salt thereof (hereinafter sometimes referred to as the polypeptide of the present invention), a method for manufacturing the same and utility thereof are described below in more detail.
  • the polypeptide of the present invention may be any polypeptide so long as it is derived from any tissues (e.g., pituitary gland, pancreas, brain, kidney, liver, genital gland, thyroid gland, gall bladder, spinal cord, adrenal, skin, muscle, lung, digestive tract, blood vessel, heart, etc.), cells or the like of human and other warm-blooded animals (e.g., guinea pig, rat, mouse, swine, sheep, bovine, monkey, etc.) and has the same or substantially the same amino acid sequence as the amino acid sequence shown by SEQ ID NO:7.
  • tissues e.g., pituitary gland, pancreas, brain, kidney, liver, genital gland, thyroid gland, gall bladder, spinal cord, adrenal, skin, muscle, lung, digestive tract, blood vessel, heart, etc.
  • cells or the like of human and other warm-blooded animals e.g., guinea pig, rat, mouse, swine, sheep, bovine,
  • polypeptide of the present invention examples include, in addition to the polypeptide containing the amino acid sequence shown by SEQ ID NO:7, a polypeptide which has substantially equivalent activity to the polypeptide containing the amino acid sequence shown by SEQ ID NO:7 (e.g., a polypeptide containing the amino acid sequence shown by SEQ ID NO:8 or SEQ ID NO:21, etc.) and the like.
  • substantially equivalent activity examples include a receptor-binding activity, a signal transduction activity, and the like.
  • the term substantially equivalent is used to mean that the nature of the receptor binding activity, etc. is equivalent. Therefore, differences in degree such as a level of the receptor binding activity and quantitative factors such as a molecular weight of the polypeptide may be present and allowable.
  • polypeptides preferred are a polypeptide containing the amino acid sequence shown by SEQ ID NO: 7 in which the third amino acid (Thr) from the N-terminus is substituted with Pro (SEQ ID NO:8) and a polypeptide containing the amino acid sequence shown by SEQ ID NO: 7 in which the third amino acid (Thr) from the N-terminus is substituted with Ser (SEQ ID NO: 21) and the like.
  • the polypeptides are represented in accordance with the conventional way of describing peptides, that is, the N-terminus (amino terminus) at the left hand and the C-terminus (carboxyl terminus) at the right hand.
  • the C-terminus is usually in the form of a carboxyl group (-COOH) or a carboxylate (-COO - ) but may be in the form of an amide (-CONH 2 ) or an ester (-COOR).
  • ester group shown by R examples include a C 1-6 alkyl group such as methyl, ethyl, n-propyl, isopropyl, n-butyl, etc.; a C 3-8 cycloalkyl group such as cyclopentyl, cyclohexyl, etc.; a C 6-12 aryl group such as phenyl, ⁇ -naphthyl, etc.; a phenyl-C 1-2 alkyl group, e.g., benzyl, phenethyl, benzhydryl, etc.; a C 7-14 aralkyl such as an ⁇ -naphthyl-C 1-2 alkyl group, e.g., ⁇ -naphthylmethyl, etc.; and the like.
  • pivaloyloxymethyl or the like which is used widely as an ester for oral administration may also be used.
  • the polypeptide of the present invention may be used in the form of salts with physiologically acceptable bases (e.g., alkali metal salts) or acids (e.g., inorganic acids or organic acids), preferably in the form of physiologically acceptable acid addition salts.
  • physiologically acceptable bases e.g., alkali metal salts
  • acids e.g., inorganic acids or organic acids
  • physiologically acceptable acid addition salts examples include salts with inorganic acids (e.g., hydrochloric acid, phosphoric acid, hydrobromic acid, sulfuric acid), salts with organic acids (e.g., acetic acid, formic acid, propionic acid, fumaric acid, maleic acid, succinic acid, tartaric acid, citric acid, malic acid, oxalic acid, benzoic acid, methanesulfonic acid, benzenesulfonic acid) and the like.
  • inorganic acids e.g., hydrochloric acid, phosphoric acid, hydro
  • polypeptide of the present invention or salts thereof may be manufactured by a method used to purify a polypeptide from human or other warm-blooded animal cells or tissues described above.
  • the polypeptide of the present invention or salts thereof may also be manufactured by the methods for synthesizing polypeptides, which will be described hereinafter, or by modified methods.
  • the polypeptide of the present invention or its salts may also be manufactured by culturing a transformant containing a DNA encoding the polypeptide, as will also be later described.
  • polypeptide or salts thereof are manufactured from human or warm-blooded animal tissues or cells
  • human or warm-blooded animal tissues or cells are homogenized, then extracted with an acid, an organic solvent or the like, and the extract is isolated and purified by means of salting-out, dialysis, gel filtration, or a combination of chromatography techniques such as reverse phase chromatography, ion exchange chromatography, affinity chromatography, and the like.
  • the polypeptide of the present invention can be manufactured by publicly known methods for polypeptide synthesis or by cleaving a polypeptide containing the polypeptide of the present invention with an appropriate peptidase.
  • peptide synthesis methods for example, either solid phase synthesis method or liquid phase synthesis method may be used. That is, partial peptides or amino acids that may compose the polypeptide of the present invention are condensed with the residual portion.
  • the desired peptide can be obtained by removing the protecting groups.
  • the publicly known condensation and protecting group removal methods include the methods described in 1)-5) below.
  • the polypeptide of the present invention may be purified and isolated by a combination of conventional purification methods such as solvent extraction, distillation, column chromatography, liquid chromatography and recrystallization.
  • the polypeptide obtained by the above methods is in a free form, the polypeptide can be converted into an appropriate salt by a publicly known method; when the polypeptide is obtained in a salt form, it can be converted into a free form by a publicly known method.
  • resins for peptide synthesis that are suitable for amide formation may be used.
  • resins include chloromethyl resin, hydroxymethyl resin, benzhydrylamine resin, aminomethyl resin, 4-benzyloxybenzyl alcohol resin, 4-methylbenzhydrylamine resin, PAM resin, 4-hydroxymethylmethylphenyl acetoamidemethyl resin, polyacrylamide resin, 4-(2',4'-dimethoxyphenyl-hydroxymethyl)phenoxy resin, 4-(2',4'-dimethoxyphenyl-Fmoc-aminoethyl) phenoxy resin, etc.
  • amino acids in which ⁇ -amino groups and functional groups on the side chains are appropriately protected are condensed on the resin, in accordance with the order of the sequence of the objective peptide, by various condensation methods publicly known in the art.
  • the peptide is excised from the resin and at the same time, the protecting groups are removed.
  • intramolecular disulfide bond-forming reaction is performed in a highly diluted solution to obtain the objective polypeptide.
  • carbodiimides are particularly preferably employed.
  • carbodiimides include DCC, N,N'-diisopropylcarbodiimide, N-ethyl-N'-(3-dimethylaminopropyl)carbodiimide, etc.
  • the protected amino acids in combination with a racemization inhibitor e.g., HOBt, HOOBt
  • a racemization inhibitor e.g., HOBt, HOOBt
  • the protected amino acids are previously activated in the form of symmetric acid anhydrides, HOBt esters or HOOBt esters, followed by adding the thus activated protected amino acids to the resin.
  • Solvents suitable for use to activate the protected amino acids or condense with the resin may be appropriately chosen from solvents that are known to be usable for peptide condensation reactions.
  • solvents examples include acid amides such as N,N-dimethylformamide, N,N-dimethylacetoamide, N-methylpyrrolidone, etc.; halogenated hydrocarbons such as methylene chloride, chloroform, etc.; alcohols such as trifluoroethanol, etc.; sulfoxides such as dimethylsulfoxide, etc.; tertiary amines such as pyridine, etc.; ethers such as dioxane, tetrahydrofuran, etc.; nitriles such as acetonitrile, propionitrile, etc.; esters such as methyl acetate, ethyl acetate, etc.; and suitable mixtures of these solvents.
  • acid amides such as N,N-dimethylformamide, N,N-dimethylacetoamide, N-methylpyrrolidone, etc.
  • halogenated hydrocarbons such as methylene chloride, chloroform, etc.
  • the reaction temperature is appropriately chosen from the range known to be applicable to peptide binding reactions and is usually selected in the range of approximately -20°C to 50°C.
  • the activated amino acid derivatives are used generally in an excess of 1.5 to 4 times.
  • the condensation is tested using the ninhydrin reaction; when the test reveals that the condensation is insufficient, the condensation can be completed by repeating the condensation reaction without removal of the protecting groups. When the condensation is still insufficient even after repeating the reaction, unreacted amino acids are acetylated with acetic anhydride or acetylimidazole to cancel any possible adverse affect on the subsequent reaction.
  • Examples of the protecting groups used to protect the starting amino groups include Z, Boc, t-pentyloxycarbonyl, isobornyloxycarbonyl, 4-methoxybenzyloxycarbonyl, Cl-Z, Br-Z, adamantyloxycarbonyl, trifluoroacetyl, phthaloyl, formyl, 2-nitrophenylsulphenyl, diphenylphosphinothioyl, Fmoc, etc.
  • Examples of the protecting group R for a carboxyl group include, in addition to those examples for the C 1-6 alkyl group, the C 3-8 cycloalkyl and the C 7 .
  • the hydroxyl group of serine and threonine can be protected through, for example, their esterification or etherification.
  • groups appropriately used for the esterification include a lower alkanoyl group such as acetyl, etc., an aroyl group such as benzoyl group, and a group derived from carbon such as benzyloxycarbonyl group and ethoxycarbonyl group.
  • groups appropriately used for the etherification include benzyl group, tetrahydropyranyl group, t-butyl group, etc.
  • Examples of groups for protecting the phenolic hydroxyl group of tyrosine include Bzl, Cl 2 -Bz1, 2-nitrobenzyl, Br-Z, t-butyl, etc.
  • Examples of groups used to protect the imidazole moiety of histidine include Tos, 4-methoxy-2,3,6-trimethylbenzenesulfonyl, DNP, benzyloxymethyl, Bum, Boc, Trt, Fmoc, etc.
  • Examples of the activated carboxyl groups in the starting materials include the corresponding acid anhydrides, azides, activated esters [esters with alcohols (e.g., pentachlorophenol, 2,4,5-trichlorophenol, 2,4-dinitrophenol, cyanomethyl alcohol, p-nitrophenol, HONB, N-hydroxysuccimide, N-hydroxyphthalimide, HOBt)].
  • activated esters esters with alcohols (e.g., pentachlorophenol, 2,4,5-trichlorophenol, 2,4-dinitrophenol, cyanomethyl alcohol, p-nitrophenol, HONB, N-hydroxysuccimide, N-hydroxyphthalimide, HOBt)].
  • activated amino groups in the starting material the corresponding phosphoric amides are employed.
  • a cation scavenger such as anisole, phenol, thioanisole, m-cresol, p-cresol, dimethyl sulfide, 1,4-butanedithiol or 1,2-ethanedithiol.
  • 2,4-dinitrophenyl group known as a protecting group for the imidazole of histidine is removed by a treatment with thiophenol.
  • Formyl group used as a protecting group of the indole of tryptophan is eliminated by the aforesaid acid treatment in the presence of 1,2-ethanedithiol or 1,4-butanedithiol, as well as by a treatment with an alkali such as a dilute sodium hydroxide solution, dilute ammonia, etc.
  • Protection of functional groups that should not be involved in the reaction of the starting materials, protecting groups, elimination of the protecting groups and activation of functional groups involved in the reaction may be appropriately selected from publicly known groups and publicly known means.
  • the ⁇ -carboxyl group of the carboxyl terminal amino acid is first amidated; the peptide chain is then extended from the amino group side to a desired length. Thereafter, a peptide in which only the protecting group of the N-terminal ⁇ -amino group has been eliminated from the peptide and a peptide in which only the protecting group of the C-terminal carboxyl group has been eliminated are manufactured.
  • the two peptides are condensed in a mixture of the solvents described above. The details of the condensation reaction are the same as described above.
  • all the protecting groups are eliminated by the method described above to give the desired crude polypeptide.
  • This crude polypeptide is purified by various known purification means. Lyophilization of the major fraction gives the amide of the desired polypeptide.
  • esterified polypeptide for example, the ⁇ -carboxyl group of the carboxy terminal amino acid is condensed with a desired alcohol to prepare the amino acid ester, which is followed by procedure similar to the preparation of the amidated polypeptide above to give the ester of the desired polypeptide.
  • Any polypeptide is usable as the polypeptide of the present invention so long as it contains the same or substantially the same amino acid sequence as that represented by SEQ ID NO:7 described above and possesses the same activities as those of the polypeptide, e.g., activities for regulating central nervous functions, circulatory functions, heart functions, renal functions, urinary functions, or sensory functions, etc.
  • a polypeptide is, for example, a peptide having the amino acid sequence shown by SEQ ID NO:8 or SEQ ID NO:21.
  • polypeptide of the present invention can be used as an antigen for preparing an antibody to the polypeptide.
  • a polypeptide that can be used as the antigen partial peptides such as N-terminal peptide, C-terminal peptide, a peptide at the central part, etc. of the polypeptide in the present invention may be employed as well, in addition to the polypeptides of the present invention described above.
  • the partial peptide that can be employed may be a peptide containing each of the individual domains or a peptide containing a plurality of domains concurrently.
  • the C-terminus may also be in the form of an amide (-CONH 2 ) or an ester (-COOR).
  • amides of the partial peptide may also be included in the partial peptide of the present invention.
  • esters are those of the C-terminal esters described above, and the like.
  • polypeptide of the present invention or its partial peptide may be a fused protein with a protein, which functions or properties are well known.
  • salts of the partial peptide which can be used for the polypeptide of the present invention are the same as those of the aforesaid polypeptide salts.
  • the partial peptide of the polypeptide of the present invention, its amide or ester, or a salt thereof may be manufactured by the same synthesis method as in the polypeptide of the present invention described above, or by cleaving the polypeptide of the present invention with an appropriate peptidase.
  • the DNA encoding the polypeptide of the present invention may be any DNA so long as it contains a DNA encoding a polypeptide containing the same or substantially the same amino acid sequence represented by SEQ ID NO:7.
  • the DNA may be any one of genomic DNA, genomic DNA library, cDNA derived from the cells or tissues described above, cDNA library derived from the cells or tissues described above and synthetic DNA.
  • the vector used for the library may be any of bacteriophage, plasmid, cosmid, phagemid and the like.
  • the DNA can be directly amplified by reverse transcriptase polymerase chain reaction (hereinafter abbreviated as RT-PCR technique) using an RNA fraction prepared from the cells or tissues described above.
  • RT-PCR technique reverse transcriptase polymerase chain reaction
  • the polypeptide containing the same or substantially the same amino acid sequence as that shown by SEQ ID NO:7 includes the amino acid sequence shown by SEQ ID NO:8 or SEQ ID NO:21, as described above.
  • the DNA containing the polypeptide-encoding DNA containing the amino acid sequence shown by SEQ ID NO:8 is, for example, a DNA containing a DNA having the base sequence shown by SEQ ID NO:27, and the like
  • the DNA containing the polypeptide-encoding DNA containing the amino acid sequence shown by SEQ ID NO:21 is, e.g., a DNA containing a DNA having the base sequence shown by SEQ ID NO:28.
  • Examples of the DNA containing a DNA encoding the polypeptide having the same or substantially the same amino acid sequence as the amino acid sequence shown by SEQ ID NO:7 are a DNA containing a DNA having a base sequence with homology of at least about 80%, preferably at least about 90%, more preferably at least about 95%, and most preferably at least about 98%, to the base sequence shown by SEQ ID NO:27 or SEQ ID NO:28.
  • Examples of the DNA containing a DNA encoding the polypeptide having the same or substantially the same amino acid sequence as the amino acid sequence shown by SEQ ID NO:7 include DNAs containing (i) the base sequence represented by SEQ ID NO:27 or SEQ ID NO:28, of which at least 1 or 2 (preferably 1 to 30, more preferably 1 to 10 and most preferably (1 or 2)) bases are deleted; (ii) the base sequence represented by SEQ ID NO:27 or SEQ ID NO:28, to which at least 1 or 2 (preferably 1 to 30, more preferably 1 to 10 and most preferably (1 or 2)) bases are added; (iii) the base sequence represented by SEQ ID NO:27 or SEQ ID NO:28, in which at least 1 or 2 (preferably 1 to 30, more preferably 1 to 10 and most preferably (1 or 2)) bases are inserted; (iv) in the amino acid sequence represented by SEQ ID NO:27 or SEQ ID NO:28, the base sequence in which at least 1 or 2 (preferably 1 to 30, more preferably 1 to 10 and most preferably
  • a mammal-derived DNA that is hybridizable under stringent conditions to the sequence possessed by a DNA containing a DNA capable of binding to a receptor protein containing the same or substantially the same amino acid sequence as the amino acid sequence represented by SEQ ID NO:7; (2) a DNA that is not hybridizable due to degeneracy of genetic code to the sequence possessed by the DNA containing a DNA capable of binding to a receptor protein containing the same or substantially the same amino acid sequence as the amino acid sequence represented by SEQ ID NO:7 and the sequence defined in (1) but encoding a polypeptide having the same amino acid sequence, and the like.
  • the hybridization can be carried out by publicly known methods or by a modified known methods.
  • DNA hybridizable to the sequence possessed by a DNA containing a DNA encoding a polypeptide containing the same or substantially the same amino acid sequence as the amino acid sequence represented by SEQ ID NO:7 there are employed, e.g., a DNA containing a base sequence having homology of at least about 70%, preferably at least about 80%, more preferably at least about 90%, and most preferably at least about 95%, to the base sequence shown by SEQ ID NO:27 or SEQ ID NO:28, and the like.
  • DNA fragments containing a partial base sequence of the DNA encoding the polypeptide containing 1) the amino acid sequence shown by SEQ ID NO:7, 2) the amino acid sequence shown by SEQ ID NO:8, 3) the amino acid sequence shown by SEQ ID NO:21, etc. are also preferably used as probes for DNA detection.
  • DNA encoding the polypeptide of the present invention may also be manufactured by the following genetic engineering techniques.
  • the DNA may be either amplified from the DNA library, etc. supra by publicly known PCR using synthetic DNA primers containing a partial base sequence of the polypeptide of the present invention or, the DNA inserted into an appropriate vector can be selected by hybridization with a labeled DNA fragment or synthetic DNA that encodes a part or entire region of the polypeptide of the present invention.
  • the hybridization can be carried out, for example, according to the method described in Molecular Cloning (2nd ed.; J. Sambrook et al., Cold Spring Harbor Lab. Press, 1989). When a commercially available library is used, the hybridization may also be performed in accordance with the protocol described in the attached instructions.
  • the cloned DNA encoding the polypeptide of the present invention can be used as it is, depending upon purpose or, if desired, after digestion with a restriction enzyme or after addition of a linker thereto.
  • the DNA may contain ATG as a translation initiation codon at the 5' end thereof and TAA, TGA or TAG as a translation termination codon at the 3' end thereof. These translation initiation and termination codons may also be added by using an appropriate synthetic DNA adapter.
  • the expression vector of the polypeptide of the present invention can be manufactured, for example, by (a) excising the desired DNA fragment from the DNA encoding the polypeptide of the present invention, (b) and then ligating the DNA fragment to an appropriate expression vector downstream of a promoter in the vector.
  • Examples of the vector include plasmids derived form E. coli (e.g., pBR322, pBR325, pUC12, pUC13), plasmids derived from Bacillus subtilis (e.g., pUB110, pTP5, pC194), plasmids derived from yeast (e.g., pSH19, pSH15), bacteriophages such as ⁇ phage, etc., animal viruses such as retrovirus, vaccinia virus, baculovirus, etc.
  • E. coli e.g., pBR322, pBR325, pUC12, pUC13
  • Bacillus subtilis e.g., pUB110, pTP5, pC194
  • yeast e.g., pSH19, pSH15
  • bacteriophages such as ⁇ phage, etc.
  • animal viruses such as retrovirus, vaccinia virus, baculovirus, etc
  • the promoter used in the present invention may be any promoter if it matches well with a host to be used for gene expression.
  • SV40-derived promoter When an animal cell is used as a host for transformation, there may be utilized SV40-derived promoter, retrovirus promoter, metallothionein promoter, heat shock promoter, cytomegalovirus promoter, SR ⁇ promoter, etc.
  • the host is bacteria of the genus Escherichia
  • preferred examples of the promoter include trp promoter, T7 promoter, lac promoter, recA promoter, ⁇ PL promoter, lpp promoter, etc.
  • preferred example of the promoter are SPO1 promoter, SPO2 promoter and penP promoter.
  • promoter When yeast is used as the host, preferred examples of the promoter are PHO5 promoter, PGK promoter, GAP promoter, ADH1 promoter, GAL promoter, etc. When insect cells are used as the host, preferred examples of the promoter include polyhedrin prompter, P10 promoter, etc.
  • the expression vector may further optionally contain an enhancer, a splicing signal, a polyA addition signal, a selection marker, SV40 replication origin (hereinafter sometimes abbreviated as SV40ori), etc.
  • the selection marker include dihydrofolate reductase (hereinafter sometimes abbreviated as dhfr) gene [methotrexate (MTX) resistance], ampicillin resistant gene (hereinafter sometimes abbreviated as Amp r ), neomycin resistant gene (hereinafter sometimes abbreviated as Neo, G418 resistance), etc.
  • dhfr dihydrofolate reductase
  • MTX metalhotrexate
  • Amp r ampicillin resistant gene
  • Neo neomycin resistant gene
  • a signal sequence that matches with a host is added to the N-terminal side of the polypeptide or its partial peptide.
  • the signal sequence that can be utilized are phoA signal sequence, OmpA signal sequence, etc. when bacteria of the genus Escherichia are used as the host; ⁇ -amylase signal sequence, subtilisin signal sequence, etc. when bacteria of the genus Bacillus are used as the host; mating factor ⁇ (MF ⁇ ) signal sequence, invertase signal sequence, etc. when yeast is used as the host; and insulin signal sequence, ⁇ -interferon signal sequence, antibody molecule signal sequence, etc. when animal cells are used as the host, respectively.
  • transformants can be prepared.
  • Examples of the host which may be employed, are bacteria belonging to the genus Escherichia, bacteria belonging to the genus Bacillus, yeast, insects or insect cells and animal cells, etc.
  • bacteria belonging to the genus Escherichia include Escherichia coli K12 DH1 (Proc. Natl. Acad. Sci. U.S.A., 60, 160 (1968)), JM103 (Nucleic Acids Research, 9, 309 (1981)), JA221 (Journal of Molecular Biology, 120, 517 (1978)), HB101 (Journal of Molecular Biology, 41, 459 (1969)), C600 (Genetics, 39, 440 (1954)), etc.
  • Bacillus subtilis MI114 Gene, 24, 255 (1983)
  • 207-21 Journal of Biochemistry, 95, 87 (1984)
  • yeast examples include Saccharomyces cereviseae AH22, AH22R - , NA87-11A, DKD-5D, 20B-12, etc.
  • a larva of Bombyx mori can be used (Maeda et al., Nature, 315, 592 (1985)).
  • insect cells examples include, for the virus AcNPV, Spodoptera frugiperda cell (Sf cell), MG1 cell derived from mid-intestine of Trichoplusia ni, High FiveTM cell derived from egg of Trichoplusia ni, cells derived from Mamestra brassicae, cells derived from Estigmena acrea, etc.; and for the virus BmNPV, Bombyx mori N cell (BmN cell), etc. is used.
  • Sf cell which can be used are Sf9 cell (ATCC CRL1711) and Sf21 cell (both cells are described in Vaughn, J. L. et al., In vitro, 13, 213-217 (1977)), etc.
  • animal cells examples include monkey cell COS-7, Vero cell, Chinese hamster cell CHO, DHFR gene deficient Chinese hamster cell CHO (dhfr - CHO cell), mouse L cell, mouse 3T3 cell, mouse myeloma cell, human HEK293 cell, human FL cell, 293 cell, C127 cell, BALB3T3 cell, Sp-2/O cell, etc.
  • Bacteria belonging to the genus Escherichia can be transformed, for example, by the method described in Proc. Natl. Acad. Sci. U.S.A., 69, 2110 (1972) or Gene, 17, 107 (1982), etc.
  • Bacteria belonging to the genus Bacillus can be transformed, for example, by the method described in Molecular & General Genetics, 168, 111 (1979), etc.
  • Yeast can be transformed, for example, by the method described in Proc. Natl. Acad. Sci. U.S.A., 75, 1929 (1978).
  • Insect cells or insects can be transformed, for example, according to the method described in Bio/Technology, 6, 47-55(1988), etc.
  • Animal cells can be transformed, for example, according to the method described in Virology, 52, 456 (1973).
  • Methods for introducing the expression vectors into the cells include, for example, the lipofection method [Felgner, P. L., et al., Proceedings of the National Academy of Sciences of the United States of America, 84, 7413 (1987)], the calcium phosphate method [Graham, F. L. and van der Eb, A., J. Virology, 52, 456-467 (1973)], the electroporation method [Nuemann, E. et al., EMBO J., 1, 841-845 (1982)], etc.
  • transformants transformed with the expression vector containing the DNA encoding the polypeptide of the present invention are obtained.
  • a method of selecting the cells by clone selection in which the aforesaid expression vectors transfected to animal cells are introduced into chromosomes. Specifically, transformants are selected using as an index the selection marker described above. Further by repeated clone selections on the transformants thus obtained using the selection marker, stable animal cell line capable of highly expressing the polypeptide of the present invention can be obtained.
  • cultivation can be performed by gradually increasing a level of MTX, resistant cells are selected thereby to amplify the DNA encoding the polypeptide of the present invention or its partial peptide in the cells together with the dhfr gene.
  • the animal cell line of higher expression can be obtained.
  • the transformant described above is cultivated under conditions that the DNA encoding the polypeptide of the present invention can express, to produce and accumulate the polypeptide of the present invention.
  • the polypeptide of the present invention can be manufactured.
  • the transformant can be appropriately cultivated in a liquid medium which contains materials required for growth of the transformant, such as carbon sources, nitrogen sources, inorganic materials, etc.
  • materials required for growth of the transformant such as carbon sources, nitrogen sources, inorganic materials, etc.
  • the carbon sources include glucose, dextrin, soluble starch, sucrose, etc.
  • the nitrogen sources include inorganic or organic materials such as ammonium salts, nitrates, corn steep liquor, peptone, casein, meat extract, soybean cake, potato extract, etc.
  • the inorganics are calcium chloride, sodium dihydrogenphosphate, magnesium chloride, etc.
  • yeast, vitamins, growth promoting factors, etc. may also be added to the medium.
  • pH of the medium is adjusted to about 5 to about 8.
  • a preferred example of the medium for cultivation of the bacteria belonging to the genus Escherichia is M9 medium supplemented with glucose and Casamino acids (Miller, Journal of Experiments in Molecular Genetics, 431-433, Cold Spring Harbor Laboratory, New York, 1972). If necessary and desired, a chemical such as 3 ⁇ -indolylacrylic acid can be added to the medium thereby to activate the promoter efficiently.
  • the transformant is usually cultivated at about 15 to about 43°C for about 3 hours to about 24 hours. If necessary and desired, the culture may be aerated or agitated.
  • the transformant is cultivated generally at about 30°C to about 40°C for about 6 hours to about 24 hours. If necessary and desired, the culture can be aerated or agitated.
  • the transformant is cultivated, for example, in Burkholder's minimal medium [Bostian, K. L. et al., Proc. Natl. Acad. Sci. U.S.A., 77, 4505 (1980)] or in SD medium [Bitter, G. A. et al., Proc. Natl. Acad. Sci. U.S.A., 81, 5330 (1984)] supplemented with 0.5% Casamino acids.
  • pH of the medium is adjusted to about 5 to about 8.
  • the transformant is cultivated at about 20°C to about 35°C for about 24 hours to about 72 hours. If necessary and desired, the culture can be aerated or agitated.
  • the transformant is cultivated in, for example, Grace's Insect Medium (Grace, T. C. C., Nature, 195, 788 (1962)) to which an appropriate additive such as immobilized 10% bovine serum is added.
  • pH of the medium is adjusted to about 6.2 to about 6.4.
  • the transformant is cultivated at about 27°C for about 3 days to about 5 days and, if necessary and desired, the culture can be aerated or agitated.
  • the transformant is cultivated in, for example, MEM medium containing about 5% to about 20% fetal bovine serum [Science, 122, 501 (1952)], DMEM medium [Virology, 8, 396 (1959)], RPMI 1640 medium [The Journal of the American Medical Association, 199, 519 (1967)], 199 medium [Proceeding of the Society for the Biological Medicine, 73, 1 (1950)], etc.
  • pH of the medium is adjusted to about 6 to about 8.
  • the transformant is usually cultivated at about 30°C to about 40°C for about 15 hours to about 60 hours and, if necessary and desired, the culture can be aerated or agitated.
  • CHO(dhfr') cell and the dhfr gene are employed as the selection markers, it is preferred to use substantially thymidine-free DMEM supplemented with dialyzed bovine fetal serum.
  • the transformant or cell is collected by a publicly known method and suspended in an appropriate buffer.
  • the transformant or cell is then disrupted by publicly known methods such as ultrasonication, a treatment with lysozyme and/or freeze-thaw cycling, followed by centrifugation, filtration, etc.
  • the buffer used for the procedures may contain a protein modifier such as urea or guanidine hydrochloride, or a surfactant such as Triton X-100 (trademark, hereinafter sometimes abbreviated as TM), etc.
  • the supernatant When the polypeptide is secreted in the culture broth, after completion of the cultivation the supernatant can be separated from the transformant or cell to collect the supernatant by a publicly known method.
  • the polypeptide of the present invention when the polypeptide of the present invention thus obtained is in a free form, it can be converted into the salt by publicly known methods or modifications thereof.
  • the polypeptide when the polypeptide is obtained in a salt form, it can be converted into the free form or a different salt form by publicly known methods or modifications thereof.
  • polypeptide of the present invention produced by the recombinant can be treated, prior to or after the purification, with an appropriate protein modifying enzyme so that the polypeptide can be suitably modified or partially removed.
  • protein modifying enzyme include trypsin, chymotrypsin, arginyl endopeptidase, protein kinase, glycosidase and the like.
  • the activity of the thus formed polypeptide of the present invention can be determined by an enzyme immunoassay using a specific antibody, or the like.
  • the DNA encoding the polypeptide of the present invention or the polypeptide of the present invention can be used: (1) for the survey of physiological activities that the polypeptide of the present invention possesses, (2) for preparing synthetic oligonucleotide probes or primers for PCR, (3) for acquiring DNAs encoding ligands to SENR or precursor proteins, (4) for the development of the receptor-binding assay system using the expression system of recombinant receptor protein and screening of a candidate drug, (5) for acquiring antibodies and antisera, (6) for the development of diagnostic agents using DNAs, RNAs, antibodies or antisera, (7) for the development of drugs such as agents for regulating central nervous functions, circulatory functions, heart functions, renal functions, urinary functions, sensory functions, etc., (8) for gene therapy, and the like.
  • polypeptide of the present invention or the DNA encoding the same When the polypeptide of the present invention or the DNA encoding the same is used as the drugs described above, its pharmaceutical preparations can be prepared in a conventional manner.
  • the polypeptide of the present invention or the DNA encoding the same can be used orally, for example, in the form of tablets with sugar coating or enteric coating, if necessary and desired, capsules, elixirs, microcapsules etc., or parenterally in the form of injectable preparations such as a sterile solution, a suspension, etc. in water or with other pharmaceutically acceptable liquid.
  • injectable preparations such as a sterile solution, a suspension, etc. in water or with other pharmaceutically acceptable liquid.
  • These preparations can be manufactured by mixing the compound described above with a physiologically acceptable carrier, a flavoring agent, an excipient, a vehicle, an antiseptic agent, a stabilizer, a binder, etc. in a unit dosage form required in a generally accepted manner that is
  • the DNA of the present invention is used, the DNA is inserted into an appropriate vector such as retrovirus vector, adenovirus vector, adenovirus-associated virus vector, etc. and then administered in a conventional manner.
  • an appropriate vector such as retrovirus vector, adenovirus vector, adenovirus-associated virus vector, etc.
  • a sterile composition for injection may be formulated according to a conventional manner used to make pharmaceutical compositions, e.g., by dissolving or suspending the active ingredients in a vehicle such as water for injection with a naturally occurring vegetable oil such as sesame oil, coconut oil, etc. to prepare the pharmaceutical composition.
  • an aqueous medium for injection examples include physiological saline and an isotonic solution containing glucose and other auxiliary agents (e.g., D-sorbitol, D-mannitol, sodium chloride, etc.) and may be used in combination with an appropriate dissolution aid such as an alcohol (e.g., ethanol), a polyalcohol (e.g., propylene glycol and polyethylene glycol), a nonionic surfactant (e.g., polysorbate 80 (TM) and HCO-50), etc.
  • an alcohol e.g., ethanol
  • a polyalcohol e.g., propylene glycol and polyethylene glycol
  • TM polysorbate 80
  • HCO-50 nonionic surfactant
  • the oily medium examples include sesame oil, soybean oil, etc., which may also be used in combination with a dissolution aid such as benzyl benzoate, benzyl alcohol, etc.
  • the preparation can be administered to, e.g., human or other mammals (e.g., mouse, rat, guinea pig, rabbit, sheep, swine, bovine, cat, dog, monkey, etc.).
  • human or other mammals e.g., mouse, rat, guinea pig, rabbit, sheep, swine, bovine, cat, dog, monkey, etc.
  • the dose of the polypeptide of the present invention or the DNA encoding the same varies depending on condition, etc.
  • the dose is normally about 0.1 mg to about 100 mg, preferably about 1.0 to about 50 mg, and more preferably about 1.0 to about 20 mg per day for the adult patient with heart failure (as 60 kg body weight).
  • the single dose varies depending on subject to be administered, target organ, condition, method for administration, etc. but it is advantageous to administer the active ingredient intravenously at a daily dose of about 0.01 to about 30 mg, preferably about 0.1 to about 20 mg, and more preferably about 0.1 to about 10 mg for the adult patient with heart failure (as 60 kg body weight).
  • the corresponding dose as converted per 60 kg body weight can be administered.
  • the precursor protein of the polypeptide of the present invention and salts thereof there are, for example, the aforesaid proteins of the present invention wherein at least 1 or 2, preferably 1 to about 200, more preferably 1 to about 120, and most preferably about 50 to about 120 amino acids are bound at the N terminus and/or the C terminus, or salts thereof.
  • the precursor protein of the present invention may be any protein so long as it is a protein derived from any tissue (e.g., pituitary gland, pancreas, brain, kidney, liver, genital gland, thyroid gland, gall bladder, spinal cord, adrenal, skin, muscle, lung, digestive tract, blood vessel, heart, etc.), any cell or the like of human or other warm-blooded animals (e.g., guinea pig, rat, mouse, swine, sheep, bovine, monkey, etc.) and has the same or substantially the same amino acid sequence as the amino acid sequence shown by SEQ ID NO:18, SEQ ID NO:19 or SEQ ID NO:29.
  • the substantially equivalent activity include the receptor binding activity, the signal transduction activity, etc.
  • the substantially equivalent activity is used to mean that the nature of the receptor binding activity, etc. is equivalent. Therefore, differences in degree such as a level of the receptor binding activity, quantitative factors such as a molecular weight of the protein may be present and allowable.
  • amino acid sequence having the same or substantially the same amino acid sequence shown by SEQ ID NO:18, SEQ ID NO:19 or SEQ ID NO:29 are amino acid sequences showing homology of at least about 50%, preferably at least about 60%, more preferably at least about 70%, much more preferably at least about 80%, further much more preferably at least about 90% and most preferably at least about 95%, to the amino acid sequence shown by SEQ ID NO:18, SEQ ID NO:19 or SEQ ID NO:29.
  • precursor protein of the polypeptide of the present invention containing the amino acid sequence shown by SEQ ID NO:8 are proteins containing the amino acid sequence shown by SEQ ID NO:18 or SEQ ID NO:19; and,
  • ester group shown by R examples include a C 1-6 alkyl group such as methyl, ethyl, n-propyl, isopropyl, n-butyl, etc.; a C 3-8 cycloalkyl group such as cyclopentyl, cyclohexyl, etc.; a C 6-12 aryl group such as phenyl, ⁇ -naphthyl, etc.; a phenyl-C 1-2 alkyl group, e.g., benzyl, phenethyl, benzhydryl, etc.; a C 7-14 aralkyl such as an ⁇ -naphthyl-C 1-2 alkyl group, e.g., ⁇ -naphthylmethyl, etc.; and the like.
  • pivaloyloxymethyl or the like which is used widely as an ester for oral administration may also be used.
  • salts of the precursor protein of the present invention are the same as the salts given for the polypeptide of the present invention described above.
  • the precursor protein of the present invention may be manufactured either by methods used to purify protein from human or other warm-blooded animal cells or tissues described above through modifications of the methods for manufacturing the polypeptide of the present invention or by modifications of methods for protein synthesis.
  • the precursor protein may also be manufactured by culturing a transformant containing a DNA encoding the precursor protein of the present invention, by modifications of the methods for manufacturing the polypeptide of the present invention described above.
  • the precursor protein of the present invention are manufactured from human or warm-blooded animal tissues or cells
  • the human or warm-blooded animal tissues or cells are homogenized, then extracted with an acid, an organic solvent or the like, and the extract is purified and separated by means of salting-out, dialysis, gel filtration, or a combination of chromatography techniques such as reverse phase chromatography, ion exchange chromatography, affinity chromatography, and the like.
  • the precursor protein of the present invention can be used as an antigen for preparing an antibody to the precursor protein.
  • a protein that can be used as the antigen partial peptides such as N-terminal peptide, C-terminal peptide, a peptide at the central part, etc. of the precursor protein of the present invention may be employed as well, in addition to the precursor proteins of the present invention described above.
  • the partial peptide may be either a peptide containing each of the individual domains or a peptide containing a plurality of domains at the same time.
  • the partial peptides of the precursor protein of the present invention, its amides or esters, or salts thereof can be manufactured by publicly known synthesis methods as in the precursor protein described above or by cleaving the precursor protein of the present invention with an appropriate peptidase.
  • the DNA encoding the precursor protein of the present invention may be any DNA so long as it contains a DNA encoding a protein containing the same or substantially the same amino acid sequence represented by SEQ ID NO:18, SEQ ID NO:19 or SEQ ID NO:29.
  • the DNA may be any one of genomic DNA, genomic DNA library, cDNA derived from the cells or tissues described above, cDNA library derived from the cells or tissues described above and synthetic DNA.
  • the vector used for the library may be any of bacteriophage, plasmid, cosmid, phagemid and the like.
  • the DNA can be directly amplified by reverse transcriptase polymerase chain reaction (hereinafter abbreviated as RT-PCR technique) using an RNA fraction prepared from the cells or tissues described above.
  • RT-PCR technique reverse transcriptase polymerase chain reaction
  • the DNA containing a DNA encoding a protein containing the same or substantially the same amino acid sequence as that shown by SEQ ID NO: 18, SEQ ID NO: 19 or SEQ ID NO: 29 includes a DNA containing a DNA having the base sequence shown by SEQ ID NO: 15, SEQ ID NO: 16, SEQ ID NO: 17 or SEQ ID NO: 30.
  • Examples of the DNA containing a DNA encoding a protein containing the same or substantially the same amino acid sequence as the amino acid sequence shown by SEQ ID NO: 18, SEQ ID NO: 19 or SEQ ID NO: 29 include DNAs carrying DNAs containing (i) the base sequence represented by SEQ ID NO: 15, SEQ ID NO: 16, SEQ ID NO: 17 or SEQ ID NO: 30, in which at least 1 or 2 (preferably about 1 to about 30, more preferably about 1 to about 10 and most preferably several (1 or 2)) bases are deleted; (ii) the base sequence represented by SEQ ID NO:15, SEQ ID NO:16, SEQ ID NO:17 or SEQ ID NO:30, to which at least 1 or 2 (preferably about 1 to about 30, more preferably about 1 to about 10 and most preferably several (1 or 2)) bases are added; (iii) the base sequence represented by SEQ ID NO:15, SEQ ID NO:16, SEQ ID NO:17 or SEQ ID NO:30, in which at least 1 or 2 (preferably about 1 to about 30, more
  • a mammal-derived DNA that is hybridizable under stringent conditions to the sequence possessed by a DNA containing a DNA encoding a protein containing the same or substantially the same amino acid sequence as the amino acid sequence represented by SEQ ID NO: 18, SEQ ID NO: 19 or SEQ ID NO: 29; and (2) a DNA that is not hybridizable due to degeneracy of genetic code to the sequence possessed by the DNA containing a DNA encoding a protein containing the same or substantially the same amino acid sequence as the amino acid sequence represented by SEQ ID NO:18, SEQ ID NO:19 or SEQ ID NO:29 and the sequence defined in (1) but encoding a protein having the same amino acid sequence, and the like.
  • the hybridization can be carried out by publicly known methods or by modifications of the known methods.
  • DNA fragments containing a partial base sequence of the DNA encoding the protein containing the same or substantially the same amino acid sequence as the amino acid sequence shown by SEQ ID NO:18, SEQ ID NO:19 or SEQ ID NO:29 are also preferably used as probes for detecting DNAs.
  • DNA encoding the precursor protein of the present invention may also be manufactured by the following genetic engineering techniques as in the polypeptide of the present invention described above.
  • the DNA encoding the precursor protein of the present invention or the precursor protein of the present invention can be used: (1) for the survey of physiological activities that the precursor protein of the present invention (or the polypeptide of the present invention) possesses, (2) for preparing synthetic oligonucleotide probes or primers for PCR, (3) for acquiring DNAs encoding the polypeptide of the present invention, (4) for the development of the receptor-binding assay system using the expression system of a recombinant receptor protein and for the screening of compounds as candidate drugs, (5) for acquiring antibodies and antisera, (6) for the development of diagnostic agents using DNAs, RNAs, antibodies or antisera, (7) for the development of drugs such as agents for regulating central nervous functions, circulatory functions, heart functions, renal functions, urinary functions, sensory functions, etc., (8) for gene therapy, and the like.
  • the receptor-binding assay system using the expression system of recombinant SENR described hereinafter can be used for screening SENR agonists or antagonists specific to a warm-blooded animal such as human. These agonists or antagonists may be employed as agents for the prevention/treatment of various diseases.
  • the SENR when an SENR is expressed in the central nervous system, the circulatory system or the cardiac, renal, urinary organ or sensory organ system, the SENR recognizes the precursor protein of the present invention or the DNA encoding the same as a ligand and is thus useful as safe and low toxic drug.
  • the precursor protein of the present invention or the DNA encoding the same participates in the actions for regulating the central nervous functions, circulatory functions, heart functions, renal functions, urinary functions, sensory functions, etc.
  • the precursor protein or DNA can be used as a therapeutic and prophylactic drug for, e.g., senile dementia, cerebrovascular dementia, dementia associated with genealogical retroplastic diseases (e.g., Alzheimer's disease, Parkinson's disease, Pick's disease, Huntington's disease, etc.), hypertension (hypotension), renal diseases (e.g., chronic renal failure, nephritis, etc.), cardiac diseases (e.g., heart failure, acute myocardial infarction, etc.), pollakiuria, urinary incontinence, deafness, dysosmia, optical abnormality, or the like.
  • senile dementia cerebrovascular dementia
  • dementia associated with genealogical retroplastic diseases e.g., Alzheimer's disease, Parkinson's disease, Pick's disease, Huntington
  • additives miscible with tablets, capsules etc. the additives given hereinabove may be employed.
  • the aqueous medium for injection may further be formulated with a buffer (e.g., phosphate buffer, sodium acetate buffer), a soothing agent (e.g., benzalkonium chloride, procaine hydrochloride, etc.), a stabilizer (e.g., human serum albumin, polyethylene glycol, etc.), a preservative (e.g., benzyl alcohol, phenol, etc.), an antioxidant, or the like.
  • a buffer e.g., phosphate buffer, sodium acetate buffer
  • a soothing agent e.g., benzalkonium chloride, procaine hydrochloride, etc.
  • a stabilizer e.g., human serum albumin, polyethylene glycol, etc.
  • a preservative e.g., benzyl alcohol, phenol, etc.
  • the preparation can be administered to, e.g., human or other mammals (e.g., mouse, rat, guinea pig, rabbit, sheep, swine, bovine, cat, dog, monkey, etc.).
  • human or other mammals e.g., mouse, rat, guinea pig, rabbit, sheep, swine, bovine, cat, dog, monkey, etc.
  • any partial peptide can be used so long as it is a partial peptide of the SENR of the present invention.
  • a part of the SENR molecule of the present invention which is exposed to the outside of a cell membrane can be used so long as it has an activity for binding to the polypeptide of the present invention.
  • SENRs or partial peptides thereof used in the present invention can be manufactured by the same methods as described in Tal, M. et al., Biochem. Biophys. Res. Commun., 209, 752-759, 1995, Marchese, A., Genomics, 29, 335-344, 1995 or EP 859052, or by modifications of these methods.
  • the SENR or its partial peptides may also be manufactured by the same methods as applied to the polypeptide of the present invention supra.
  • Examples of the salts of SENR or their partial peptides used in the present invention include those as given for the salts of the polypeptide of the present invention described above.
  • the DNA encoding the SENR or its partial peptide used in the present invention may be any DNA so long as it contains a DNA encoding the SENR or its partial peptide described above.
  • the DNA may be any one of genomic DNA, genomic DNA library, cDNA derived from the cells or tissues described above, cDNA library derived from the cells or tissues described above and synthetic DNA.
  • the vector used for library may also be any of bacteriophage, plasmid, cosmid, phagemid and the like.
  • the DNA can be directly amplified by RT-PCR technique using an RNA fraction prepared from the cells or tissues described above.
  • the DNA encoding the SENR or its partial peptide employed in the present invention may also be obtained by the same methods as described in Tal, M. et al., Biochem. Biophys. Res. Commun., 209, 752-759, 1995, Marchese, A., Genomics, 29, 335-344, 1995 or EP 859052, or by modifications of these methods.
  • the DNA encoding the polypeptide of the present invention may also be employed as a prophylactic/therapeutic agent for deficiency of the polypeptide or SENR, depending upon the activities of the polypeptide of the present invention and its precursor protein on the SENR.
  • the activity of the polypeptide or its precursor protein of the present invention can be provided sufficiently or properly for the patient, (a) by administering the DNA encoding the polypeptide of the present invention or its precursor protein to the patient to express the same, or (b) by inserting the DNA encoding the polypeptide or its precursor protein of the present invention into brain cells, etc.
  • the DNA described above is used as the prophylactic/therapeutic agent supra
  • the DNA alone is administered, or the DNA is inserted into an appropriate vector such as retrovirus vector, adenovirus vector, adenovirus-associated virus vector, etc. and then administered, by the same means as in the case of using the DNA encoding the polypeptide of the present invention, its precursor protein, or partial peptides thereof as drugs described above.
  • the polypeptide of the present invention or its precursor protein is capable of binding to the SENR or its salts or its partial peptides or salts thereof, the in vivo level of the SENR or its salts or the partial peptides of SENR or salts thereof can be quantified with a good sensitivity.
  • This method for quantification can be used in combination with, e.g., the competitive method. That is, the level of SENR or its salts, or the partial peptides of SENR or its salts in a sample fluid can be measured by bringing the sample fluid in contact with the polypeptide of the present invention or its precursor protein.
  • polypeptide of the present invention is used to mean the “polypeptide of the present invention” and “polypeptide containing the amino acid sequence shown by SEQ ID NO:22” described above
  • precursor of the polypeptide of the present invention is used to mean the “precursor of the polypeptide of the present invention” and “precursor of the polypeptide containing the amino acid sequence shown by SEQ ID NO:22” described above.
  • DNA encoding the polypeptide of the present invention is used to mean the “DNA encoding the polypeptide of the present invention” and “DNA encoding polypeptide containing the amino acid sequence shown by SEQ ID NO:22” described above
  • DNA encoding the precursor of the polypeptide of the present invention is used to mean the "DNA encoding the precursor of the polypeptide of the present invention” and "DNA encoding the precursor of the polypeptide containing the amino acid sequence shown by SEQ ID NO:22” described above.
  • the present invention provides a method for screening a compound or its salts that alter the binding property between the polypeptide of the present invention or its precursor protein and the SENR described above, which comprises comparing (i) the case that the polypeptide of the present invention or its precursor protein is brought into contact with the SENR or its salts or the partial peptide of the SENR or its salts, and (ii) the case that the polypeptide of the present invention or its precursor protein and a test compound are brought into contact with the aforesaid SENR or its salts or the partial peptide of the SENR or its salts.
  • the screening method of the present invention provides the following features.
  • the SENR used for the screening methods of the present invention may be any SENR so long as it contains the aforesaid SENR or the partial peptide of the SENR.
  • Preferred SENR includes membrane fractions from the organs of human or warm-blooded animals. Since it is very difficult to obtain human-derived organs, the SENR expressed in large quantities by use of recombinants is suitable as an SENR for the screening.
  • the cells or cell membrane fractions may be prepared by the method which will be described hereinafter.
  • the cells may be fixed using glutaraldehyde, formalin, etc.
  • the fixation can be made by a publicly known method.
  • an appropriate SENR fraction and the labeled polypeptide of the present invention or its precursor protein are employed.
  • the SENR fraction is preferably a naturally occurring SENR fraction or a recombinant SENR fraction having an activity equivalent to that of the natural SENR fraction.
  • the term "equivalent activity" is intended to mean a ligand binding activity, or the like, equivalent to that of the natural SENR fraction.
  • the labeled ligand there are used a labeled ligand, a labeled ligand analog compound, etc. For example, a ligand which is labeled with [ 3 H], [ 125 I], [ 14 C], [ 35 S], etc. can be utilized.
  • a standard receptor preparation is prepared by suspending SENR-containing cells or membrane fractions thereof in a buffer appropriate for the screening.
  • Any buffer can be used so long as it does not interfere with ligand-receptor binding, such buffers including a phosphate buffer or a Tris-HCl buffer having pH of 4 to 10 (desirably pH of 6 to 8).
  • a surfactant such as CHAPS, Tween-80TM (Kao-Atlas Inc.), digitonin, deoxycholate, etc. may optionally be added to the buffer.
  • a protease inhibitor such as PMSF, leupeptin, E-64 (manufactured by Peptide Institute, Inc.) and pepstatin may also be added.
  • a given amount (5,000 to 500,000 cpm) of the labeled polypeptide of the present invention is added to 0.01 ml to 10 ml of the receptor solution and at the same time, a test compound of 10 -10 to 10 -7 M is allowed to be co-present.
  • NBS non-specific binding
  • the test compound giving a non-specific binding (B-NSB) of, e.g., 50% or less can be selected as a candidate substance having a competitive inhibitory activity.
  • the method (4) or (5) described above for screening the compound that alters the binding property between the polypeptide of the present invention or its precursor protein and an SENR can be performed as follows.
  • the SENR-mediated cell-stimulating activities e.g., the activities that promote or suppress release of arachidonic acid, release of acetylcholine, release of intracellular Ca 2+ , intracellular cAMP production, intracellular cGMP production, inositol phosphate production, change in cell membrane potential, phosphorylation of intracellular proteins, activation of c-fos, pH reduction, etc.
  • the SENR-mediated cell-stimulating activities e.g., the activities that promote or suppress release of arachidonic acid, release of acetylcholine, release of intracellular Ca 2+ , intracellular cAMP production, intracellular cGMP production, inositol phosphate production, change in cell membrane potential, phosphorylation of intracellular proteins, activation of c-fos, pH
  • a suitable cell in which an SENR has been expressed is required.
  • a cell that the SENR of the present invention has been expressed is desirably the aforesaid recombinant SENR-expressed cell line, and the like.
  • test compound examples include a peptide, a protein, a non-peptide compound, a synthetic compound, a fermentation product, a cell extract, a plant extract, an animal tissue extract and the like.
  • the kit for screening the compound or its salt of the present invention that alter the binding property between the polypeptide of the present invention or its precursor protein and an SENR comprises an SENR or its salts, partial peptides of the SENR or their salts, SENR-containing cells or membrane fractions of the SENR-containing cells, and the polypeptide of the present invention or its precursor protein.
  • the screening kit according to the present invention comprises, for example, the following:
  • the buffers may be sterilized by filtration through a membrane filter with a 0.45 ⁇ m pore size and stored at 4°C, or may be prepared at use.
  • SENR-expressed CHO cells are subcultured at 5 x 10 5 cells/well on a 12-well plate followed by culturing at 37°C under a 5% CO 2 and 95% air for 2 days.
  • Ligand labeled with [ 3 H], [ 125 I], [ 14 C], [ 35 S], etc. The product dissolved in a suitable solvent or buffer is stored at 4°C or at -20°C, which will be diluted at use to 1 ⁇ M with a buffer for the assay.
  • polypeptide of the present invention or its precursor protein are dissolved in PBS containing 0.1% of bovine serum albumin (manufactured by Sigma) to make the volume 1 mM and then stored at -20°C.
  • Cells are cultured in a 12-well tissue culture plate to express the SENR. After washing the CHO cells twice with 1 ml of buffer for the assay, 490 ⁇ l of the buffer for assay is added to each well.
  • the compound or salts thereof obtainable by the screening methods or by the screening kit of the present invention is the compound that alters the binding property between the polypeptide of the present invention or its precursor protein and an SENR (inhibits or accelerates the binding), specifically a compound or salts thereof having an SENR-mediated cell-stimulating activity (so-called SENR agonists) or a compound having no cell-stimulating activity (so-called SENR antagonists).
  • SENR agonists SENR-mediated cell-stimulating activity
  • SENR antagonists a compound having no cell-stimulating activity
  • the compound include a peptide, a protein, a non-peptide compound, a synthetic compound, a fermentation product, etc. and the compound may be either novel or known compound.
  • the binding assay recited in the screening methods (1) through (3) supra is performed to obtain the compound that alters the binding property between the polypeptide of the present invention or its precursor protein and an SENR (especially inhibits the binding) followed by assay for the compound to determine if the compound has the SENR-mediated cell-stimulating activity described above.
  • the compound having the cell-stimulating activity or its salts are SENR agonists, whereas the compound having no such activity or its salts are SENR antagonists.
  • test compound is brought into contact with SENR-containing cells to assay the SENR-mediated cell-stimulating activity.
  • the compound having the cell-stimulating activity or its salts are SENR agonists.
  • the SENR-mediated cell-stimulating activity is measured both when a compound (e.g., the polypeptide of the present invention or its precursor protein, an SENR agonist, etc.) that activates an SENR is brought into contact with an SENR-containing cell and when the compound that activates the SENR and a test compound are brought into contact with the SENR-containing cell and comparison is made on the cell-stimulating activity between the two cases.
  • a compound e.g., the polypeptide of the present invention or its precursor protein, an SENR agonist, etc.
  • the compound that can reduce the cell-stimulating activity by the SENR-activating compound or its salts are SENR antagonists.
  • the SENR agonists exhibit activities similar to the physiological activities of the polypeptide of the present invention or its precursor protein and are thus useful as safe and low toxic drugs likewise the polypeptide of the present invention or its precursor protein.
  • the SENR antagonists can suppress the physiological activities that the polypeptide of the present invention possesses, they are useful as safe and low toxic drugs for suppressing the receptor activity.
  • the polypeptide of the present invention or its precursor protein participates in the actions for regulating the central nervous functions, circulatory functions, heart functions, renal functions, urinary functions, sensory functions, etc.
  • the SENR agonists may be used as a therapeutic and prophylactic agent for, e.g., senile dementia, cerebrovascular dementia, dementia associated with genealogical retroplastic diseases (e.g., Alzheimer's disease, Parkinson's disease, Pick's disease, Huntington's disease, etc.), hypertension (hypotension), renal diseases (e.g., chronic renal failure, nephritis, etc.), cardiac diseases (e.g., heart failure, acute myocardial infarction, etc.), pollakiuria, urinary incontinence, deafness, dysosmia, optical abnormality, or the like.
  • senile dementia cerebrovascular dementia
  • dementia associated with genealogical retroplastic diseases e.g., Alzheimer's disease, Parkinson's disease, Pick's disease, Huntington's
  • the salt of compound obtainable by using the screening methods or kits described above is preferably a pharmaceutically acceptable salt, exemplified by salts with inorganic bases, salts with organic bases, salts with inorganic acids, salts with organic acids and salts with basic or acidic amino acids, and the like.
  • salts with inorganic bases include alkali metal salts such as sodium salts, potassium salts, etc.; alkaline earth metal salts such as calcium salts, magnesium salts, etc.; and aluminum salts, ammonium salts, and the like.
  • salts with organic bases include salts with trimethylamine, triethylamine, pyridine, picoline, 2,6-lutidine, ethanolamine, diethanolamine, triethanolamine, cyclohexylamine, dicyclohexylamine, N,N'-dibenzylethylenediamine, etc.
  • salts with inorganic acids include salts with hydrochloric acid, hydrobromic acid, sulfuric acid, phosphoric acid, etc.
  • salts with organic acids include salts with formic acid, acetic acid, propionic acid, fumaric acid, oxalic acid, tartaric acid, maleic acid, citric acid, succinic acid, malic acid, methanesulfonic acid, benzenesulfonic acid, benzoic acid, etc.
  • Preferred examples of the salts with basic amino acids include salts with arginine, lysine, ornithine, etc.
  • Preferred examples of the salts with acidic amino acids include salts with aspartic acid, glutamic acid, etc.
  • polyclonal antibodies can be manufactured by the method described below.
  • a polyclonal antibody to the polypeptide of the present invention or its precursor protein can be manufactured by publicly known methods or modifications thereof.
  • the polyclonal antibody can be manufactured, for example, by producing the complex of an immunogen (a polypeptide antigen, etc.) and a carrier protein, immunizing a warm-blooded animal (e.g., mammals (e.g., rabbit, sheep, goat, rat, mouse, guinea pig, bovine, horse, swine), birds (e.g., chicken, dove, duck, goose, quail), etc.) as well as the method for manufacturing a monoclonal antibody described below, collecting the product containing the antibody to the polypeptide of the present invention from the immunized animal and then separating and purifying the antibody.
  • an immunogen a polypeptide antigen, etc.
  • a carrier protein e.g., immunizing a warm-blooded animal (e.g., mammals (e.g., rabbit, sheep, goat, rat
  • a type of the carrier protein and a mixing ratio of the carrier to the hapten may be any type and in any ratio, as long as the antibody is efficiently produced to the hapten immunized by crosslinking to the carrier.
  • bovine serum albumin, bovine thyroglobulin, Keyhole limpet hemocyanin, etc. is coupled to the hapten in a carrier-to-hapten weight ratio of approximately 0.1 to 20, preferably about 1 to about 5.
  • the condensation product is administered to warm-blooded animals either solely or together with carriers or diluents to the site that can produce the antibody by the administration.
  • complete Freund's adjuvant or incomplete Freund's adjuvant may be administered.
  • the administration is usually made once every about 2 to about 6 weeks and about 3 to about 10 times in total.
  • the polyclonal antibody titer to the polypeptide of the present invention or its precursor protein in antisera can be assayed by the same procedure as that for determining the antibody titer in hybridoma culture supernatant described hereinafter.
  • the separation and purification of the antibody can be made, following the method for the separation and purification of immunoglobulins performed as in the separation and purification of monoclonal antibodies described below.
  • the polypeptide of the present invention or its precursor protein is administered to warm-blooded animals (for example, mammals (e.g., rabbit, sheep, goat, rat, mouse, guinea pig, bovine, horse, swine), birds (e.g., chicken, dove, duck, goose, quail, etc.) and the like) either solely or together with carriers or diluents to the site where the production of antibody is possible by the administration.
  • mammals e.g., rabbit, sheep, goat, rat, mouse, guinea pig, bovine, horse, swine
  • birds e.g., chicken, dove, duck, goose, quail, etc.
  • complete Freund's adjuvants or incomplete Freund's adjuvants may be administered.
  • the administration is usually carried out once every 2 to 6 weeks and 2 to 10 times in total.
  • the aforesaid warm-blooded animal e.g., mice
  • immunized with an antigen wherein the antibody titer is noted is selected, then spleen or lymph node is collected after 2 to 5 days from the final immunization and antibody-producing cells contained therein are fused with myeloma cells to give monoclonal antibody-producing hybridomas.
  • the antibody titer in antisera may be assayed, for example, by reacting the labeled polypeptide of the present invention or its precursor protein, or partial peptides thereof, which will be described later, with the antiserum followed by assaying the activity of the labeling agent bound to the antibody.
  • the fusion may be carried out, for example, by the known method by Koehler and Milstein [(Nature, 256, 495, (1975)].
  • the fusion accelerator used include polyethylene glycol (PEG), Sendai virus, etc., of which PEG is preferably employed.
  • RPMI 1640 medium containing 1% to 20%, preferably 10% to 20% fetal bovine serum, GIT medium (Wako Pure Chemical Industries, Ltd.) containing 1% to 10% fetal bovine serum, a serum free medium for cultivation of a hybridoma (SFM-101, Nissui Seiyaku Co., Ltd.) and the like may be used for the selection and growth medium.
  • the cultivation is carried out generally at 20°C to 40°C, preferably at 37°C, for about 5 days to about 3 weeks, preferably 1 to 2 weeks, normally in 5% CO 2 .
  • the antibody titer of the culture supernatant of the hybridoma can be determined as in the assay for the antibody titer in the antisera described above.
  • the monoclonal antibody to the polypeptide of the present invention or its precursor protein can be separated and purified, as applied to conventional separation and purification of polyclonal antibodies, according to the methods for separation and purification of immunogloblins [e.g., salting-out, alcohol precipitation, isoelectric point precipitation, electrophoresis, adsorption/desorption with ion exchangers (e.g., DEAE), ultracentrifugation, gel filtration, or a specific purification method which comprises collecting only an antibody by means of an antigen-binding solid phase or, an activated adsorbent such as Protein A or Protein G and dissociating the binding to obtain the antibody].
  • immunogloblins e.g., salting-out, alcohol precipitation, isoelectric point precipitation, electrophoresis, adsorption/desorption with ion exchangers (e.g., DEAE), ultracentrifugation, gel filtration, or a specific purification method which comprises collecting only an antibody by means of an
  • the antibodies to the polypeptide of the present invention or its precursor protein manufactured by the methods (a) and (b) described above are capable of specifically recognizing the polypeptide of the present invention or its precursor protein, respectively.
  • these antibodies can be used for quantification of the polypeptide of the present invention or its precursor protein in a test sample fluid, in particular, for quantification by sandwich immunoassay.
  • the polypeptide of the present invention or its precursor protein may be assayed using the monoclonal antibody capable of recognizing the polypeptide of the present invention or its precursor protein.
  • the monoclonal antibody may also be used for detection by means of tissue staining, etc.
  • the antibody molecule per se may be used or F(ab') 2 , Fab' or Fab fractions of the antibody molecule may be used as well.
  • the assay method using the antibody of the present invention There is no particular limitation for the assay method using the antibody of the present invention.
  • any method may be used so far as it relates to a method in which the amount of antibody, antigen or antibody-antigen complex can be detected by a chemical or physical means, depending on or corresponding to the amount of antigen (e.g., the amount of the polypeptide) in a test sample fluid to be assayed, and then calculated using a standard curve prepared by a standard solution containing a known amount of the antigen.
  • the amount of antigen e.g., the amount of the polypeptide
  • a test sample fluid to be assayed e.g., the amount of the polypeptide
  • a standard curve prepared by a standard solution containing a known amount of the antigen.
  • the sandwich method which will be described later, is particularly preferred.
  • Examples of the labeling agent used in the assay method using a labeling substance are radioisotopes, enzymes, fluorescent substances and luminescent substances, etc.
  • Examples of the radioisotope are [ 125 I], [ 131 I], [ 3 H], [ 14 C], etc.
  • Preferred examples of the enzyme are those that are stable and have a high specific activity, including ⁇ -galactosidase, ⁇ -glucosidase, an alkaline phosphatase, a peroxidase and malate dehydrogenase.
  • Examples of the fluorescent substance are fluorescamine, fluorescein isothiocyanate, etc.
  • Examples of the luminescent substance are luminol, a luminol derivative, luciferin, lucigenin, etc.
  • the biotin-avidin system may be used as well for binding between an antibody or antigen and a labeling agent.
  • the carrier include insoluble polysaccharides such as agarose, dextran and cellulose; synthetic resins such as polystyrene, polyacrylamide, silicone, etc.; glass; and the like.
  • a test sample fluid is reacted with an immobilized anti-polypeptide antibody (first reaction), then reacted with another labeled anti-polypeptide antibody (second reaction) and the activity of the labeling agent on the insoluble carrier is measured, whereby the amount of the polypeptide in the test sample fluid can be quantified.
  • the first and second reactions may be carried out in a reversed order or simultaneously or they may be conducted with an interval.
  • the type of the labeling agent and the method for immobilization may be similar to those described hereinabove.
  • the antibody used for the labeled antibody and for the solid phase should be one type or one species but a mixture of two or more antibodies may also be used for the purpose of improving the measurement sensitivity, etc.
  • antibodies wherein the binding sites to the polypeptide of the present invention or its precursor protein are different from one another are preferably used as the anti-polypeptide antibody or its anti-precursor protein antibody used for the first and the second reactions.
  • the antibodies used in the first and the second reactions are those wherein, when the antibody used in the second reaction recognizes the C-terminal region of the polypeptide of the present invention or its precursor protein, the antibody recognizing the site other than the C-terminal regions, e.g., recognizing the N-terminal region, is preferably used in the first reaction.
  • liquid phase method in which a soluble antibody is used as the antibody and the B/F separation is conducted by polyethylene glycol, a second antibody to the said antibody, etc. is used; and a solid phase method in which an immobilized antibody is used as the first antibody or, a soluble antibody is used as the first antibody while an immobilized antibody is used as the second antibody.
  • the assay system for the polypeptide, its precursor protein or their partial peptides of the present invention may be constructed in addition to conditions or operations conventionally used for each of the methods, taking into account the technical consideration of one skilled in the art. For the details of such conventional technical means, a variety of reviews, reference books, etc.
  • Hiroshi Irie (ed.): “Radioimmunoassay” (published by Kodansha, 1974)]; Hiroshi Irie (ed.): “Radioimmunoassay; Second Series” (published by Kodansha, 1979); Eiji Ishikawa, et al. (ed.): “Enzyme Immunoassay” (published by Igaku Shoin, 1978); Eiji Ishikawa, et al. (ed.): “Enzyme Immunoassay” (Second Edition) (published by Igaku Shoin, 1982); Eiji Ishikawa, et al.
  • test sample fluid can be prepared from mammals to be tested (e.g., human, rabbit, sheep, goat, rat, mouse, guinea pig, bovine, horse, swine) by publicly known methods.
  • mammals to be tested e.g., human, rabbit, sheep, goat, rat, mouse, guinea pig, bovine, horse, swine
  • test sample fluid include blood, lymph, urine, etc.
  • bases, amino acids, etc. are shown by abbreviations, the codes of bases and amino acids are denoted in accordance with the IUPAC-IUB Commission on Biochemical Nomenclature or by the codes conventionally used in the art, examples of which are shown below. With respect to amino acids that may have their optical isomers, L form is presented unless otherwise indicated.
  • sequence identification numbers in the sequence listing of the specification indicate the following sequences, respectively.
  • Transformant Escherichia coli XL1 Blue/pZ1-puro2 containing the base sequence shown by SEQ ID NO:15, which was obtained in EXAMPLE 10 later described was on deposit with the Ministry of International Trade and Industry, Agency of Industrial Science and Technology, National Institute of Bioscience and Human Technology (NIBH) as the Accession Number FERM BP-6858 on August 23, 1999 and with Institute for Fermentation (IFO) as the Accession Number IFO 16271 on March 18, 1999.
  • Transformant Escherichia coli TOP10/pCR-buro containing the base sequence shown by SEQ ID NO:36, which was obtained in EXAMPLE 36 later described was on deposit with the Ministry of International Trade and Industry, Agency of Industrial Science and Technology, National Institute of Bioscience and Human Technology (NIBH) as the Accession Number FERM BP-6932 on November 8, 1999 and with Institute for Fermentation (IFO) as the Accession Number IFO 16332 on October 27, 1999.
  • rat brain-derived poly(A) + RNA (Clonetech Laboratories, Inc.) as a template
  • reverse transcription was carried out using a random primer.
  • reagents available from Takara RNA PCR ver. 2 kit were used.
  • amplification by PCR was performed, using the synthetic DNA primers shown by SEQ ID NO:1 and SEQ ID NO:2.
  • the synthetic DNA primers were constructed to amplify genes at the region to be translated into receptor proteins. In this case, recognition sites of the respective restriction enzymes were added at the 5' and 3' sides so that the base sequence recognized by restriction enzyme Sal I will be added at the 5' side of the gene and the base sequence recognized by restriction enzyme Spe I at the 3' side.
  • the reaction solution was composed of 5 ml of the cDNA template, 1 ⁇ M each of the synthetic DNA primers, 0.2 mM dNTPs, 1 mM MgCl 2 , 1 ⁇ l of KOD (King of DNA) DNA polymerase and a buffer attached to the enzyme, which were mixed together to make the total volume 50 ⁇ l.
  • KOD King of DNA
  • a cycle was set to include 94°C for 30 seconds, 59°C for 30 seconds and 74°C for 60 seconds. Totally this cycle was repeated 35 times. Amplification of DNAs was confirmed by 0.8% agarose gel electrophoresis followed by ethidium bromide staining.
  • the PCR products obtained by PCR in EXAMPLE 1 were separated by using a 0.8% low-melting temperature agarose gel. The band parts were excised from the gel with a razor blade and then homogenized. The homogenate was extracted with phenol and then with phenol/chloroform and precipitated in ethanol to recover DNAs. According to the protocol attached to PCR-ScriptTM Amp SK(+) Cloning Kit (Stratagene Co.), the recovered DNAs were subcloned into the plasmid vector, pCR-Script Amp SK(+). The recombinant vectors were introduced into Escherichia coli JM109 competent cells (Takara Shuzo Co.) to produce transformants.
  • clones having a cDNA-inserted fragment were selected in an LB agar medium supplemented with ampicillin and X-gal. Only the clones exhibiting white color were picked up with a sterilized toothpick to obtain transformant E. coli JM109/SENR. The individual clones were cultured overnight in an LB medium containing ampicillin. Plasmid DNAs were prepared using QIA prep8 mini prep (Qiagen Co.). An aliquot of the DNAs thus prepared was processed for cleavage by restriction enzymes Sal I and Spe I to confirm the size of the receptor cDNA fragment inserted.
  • the reaction for base sequencing was carried out by using a DyeDeoxy Terminator Cycle Sequence Kit (Perkin-Elmer Co.), followed by decoding with a fluorescent automatic sequencer. It was confirmed by sequence analysis that the sequences of 3 clones obtained entirely coincided with the gene sequence in the DNA sequence (Tal, M. et al., Biochem. Biophys. Res. Commun., Vol. 209, pp. 752-759 (1995)) of the full sequence-reported SENR (sensory epithelial neuropeptide-like receptor), wherein the Sal I recognition site was added at the 5' side and the Spe I recognition site was added at the 3' side (FIG. 1 and SEQ ID NO:3).
  • SENR sensor epithelial neuropeptide-like receptor
  • Plasmid Midi Kit Plasmid Midi Kit (Qiagen Co.) plasmid was prepared from clones of E. coli transformed by the plasmid encoding the rat brain-derived SENR full-length amino acid sequence, which sequence was confirmed in EXAMPLE 2, with the Sal I recognition sequence added at the 5' side and with the Spe I recognition sequence added at the 3' side.
  • the plasmid was cleaved with restriction enzymes Sal I and Spe I to excise the insert part out. After electrophoresis, the insert DNA was excised from the agarose gel with a razor blade and then homogenized. The homogenate was extracted with phenol and then with phenol/chloroform, followed by precipitation in ethanol. Thus, the insert DNA was recovered.
  • the insert DNA was added to Sal I- and Spe I-cleaved vector plasmid pAKK0-111H for animal cell expression (the same as the vector plasmid pAKK01.11G described in Hinuma, S. et al., Biochim. Biophys. Acta, Vol. 1219, pp. 251-259 (1994)) followed by ligation using T4 ligase (Takara Shuzo Co.).
  • T4 ligase T4 ligase
  • Plasmid Midi Kit Qiagen Co.
  • the plasmid DNA was introduced into CHO dhfr - cells.
  • DNA, 10 ⁇ g was co-precipitated with calcium phosphate in suspension.
  • the resulting suspension was added to a 10 cm Petri dish in which 5 ⁇ 10 5 or 1 ⁇ 10 6 CHO dhfr - cells had been seeded before 24 hours.
  • the cells were cultured in MEM ⁇ containing 10% fetal calf serum for one day. After passage, the cells were cultured in nucleic acid-free selection medium MEM ⁇ containing 10% dialyzed fetal calf serum and 68 clones of the transformant SENR-expression CHO cells, growing in the selection medium, were selected.
  • the amounts of the expressed full-length SENR receptor protein mRNAs of 68 clones from the CHO/SENR strain established in EXAMPLE 3 were measured as follows using Cytostar T Plate (Amersham Pharmacia Biotech Co.), in accordance with the protocol attached thereto. Each clone of the CHO/SENR strain was inoculated on Cytostar T Plate in 2.5 x 10 4 cells/ well. After culturing for 24 hours, the cells were fixed with 10% formalin. After 0.25% Triton X-100 was added to each well to increase cell permeability, 35 S-labeled riboprobe of SEQ ID NO:4 was added to the cells for hybridization.
  • the homogenate was centrifuged (8,000 rpm, 30 mins.) and the supernatant was taken out. After 1.4 liter of 1.0 M acetic acid was added to the precipitate, the mixture was again homogenized by a polytron. The homogenate was stirred overnight and then centrifuged (8,000 rpm, 30 mins.) to obtain the supernatant. After 2-fold volume of chilled acetone was slowly added dropwise to the supernatant at 4°C, the supernatant obtained by the first centrifugation was stirred overnight and, the supernatant obtained by the second centrifugation was stirred for 4 hours.
  • the acetone-added extract was centrifuged (8,000 rpm, 30 mins.) to remove the precipitate and acetone was evaporated off in vacuum from the supernatant, using an evaporator.
  • An equal volume of diethyl ether was added to the acetone-removed extract, the ethereal layer containing lipids was separated using a separating funnel to recover the aqueous layer. After the lipids were removed with ether, the extract was concentrated in vacuum using an evaporator to completely remove the ether.
  • CHO/SENR cells and mock CHO cells were inoculated on a 24-well plate in 5 x 10 4 cells/well. After incubation for 24 hours, [ 3 H] arachidonic acid was added to the system in 0.25 ⁇ Ci/well. Sixteen hours after the addition of [ 3 H] arachidonic acid, the cells were washed with Hanks' balanced salt solution (HBSS) supplemented with 0.05% bovine serum albumin (BSA) and 500 ⁇ l of 0.05% BSA-containing HBSS, to which 2 ⁇ l (corresponding to 2 g of the spinal cord) of the aforesaid DMSO solution of the HPLC fraction was added, was added to each well.
  • HBSS Hanks' balanced salt solution
  • BSA bovine serum albumin
  • the arachidonic acid metabolite release-promoting activity was expressed by % in terms of the amount of the [ 3 H] arachidonic acid metabolites released upon addition of the HPLC fraction (2 ⁇ l), when the amount of the [ 3 H] arachidonic acid metabolites released when 2 ⁇ l of DMSO alone was added was made 100%.
  • the arachidonic acid metabolite release-promoting activity specific to the CHO/SENR cell line was noted with the fraction #33. Since the arachidonic acid metabolite release-promoting activity noted from fractions #26 through #29 was observed in the mock CHO cells as well, the arachidonic acid metabolite release-promoting activity was not specific to the CHO/SENR cell line. The activity was expressed in terms of percentage to the amount of arachidonic acid metabolites released in the control group where DMSO alone was added.
  • HPLC fraction #33 which showed the arachidonic acid metabolite release activity to the CHO/SENR cells in EXAMPLE 5 was treated with proteolytic enzyme, pronase (Sigma, protease Type XIV (P5147)) to examine if the active substance is proteinaceous.
  • HPLC fraction (#33), 4 ⁇ l, from the spinal cord extract described above was added to 200 ⁇ l of 0.2 M ammonium acetate and 3 ⁇ l of pronase was further added thereto. After incubation at 37°C for 2 hours, the culture was boiled in boiling water for 10 minutes to inactivate the pronase. To the reaction solution was added 2 ml of distilled water containing 0.05 mg of BSA and 0.05 mg of CHAPS, followed by lyophilization.
  • pronase alone the HPLC fraction alone, and a mixture of the HPLC fraction with pronase alone after its heating were treated in a similar manner and then lyophilized.
  • Each sample fluid lyophilized was dissolved in 500 ⁇ l of 0.05% BSA-containing HBSS.
  • the solution was added to the CHO/SENR cells by the procedures described in EXAMPLE 5 followed by assay for the arachidonic acid metabolite release activity. The results are shown in FIG. 3.
  • the activity was expressed in terms of percentage to the amount of arachidonic acid metabolites released in a well charged with 500 ⁇ l of 0.05% BSA-containing HBSS. Since the active substances showing the arachidonic acid metabolite release activity on the CHO/SENR cells in the porcine spinal cord extract were completely inactivated, the substances were suspected to be proteins or peptides.
  • EXAMPLE 7 Purification of the active substances showing the arachidonic acid metabolite release activity specific to the SENR-expression CHO cells from the swine spinal cord
  • Swine spinal cord 1.0 kg (corresponding to 50 pigs), which had been purchased from Tokyo Shibaura Zoki Co. and kept under ice cooling after the spinal cord was withdrawn from swine on the day of their sacrifice, was homogenized in 10 1 of 70% acetone containing 40 mM hydrochloric acid and 1.0 M acetic acid using Polytron (20,000 rpm, 10 min). The homogenate was centrifuged (8,000 rpm, 30 mins.) and the supernatant was taken out.
  • the concentrate was filtrated through a glass fiber-made filter paper (Advantech, DP70 (90 mm ⁇ )) and a half of the filtrate was charged in a glass-made column (30 ⁇ x 240 mm) packed with C18 (YMC, YMCgel ODS-AM 120-S50). After washing with 400 ml of 1.0 M acetic acid, the column was eluted with 500 ml of 60% acetonitrile containing 0.1% trifluoroacetic acid. The eluate was concentrated in vacuum, the solvent was distilled off. The concentrate was then lyophilized. The remaining half of the filtrate was similarly treated and lyophilized.
  • the active fractions were lyophilized followed by dissolving in 1.0 ml of 10% acetonitrile containing 0.1% trifluoroacetic acid. After 0.5 ml each of the solution was passed through a diphenyl column (Separation Group, Vydac 219-TP54), elution was performed by density gradient of 26% to 31% acetonitrile containing 0.1% trifluoroacetic acid. HPLC was carried out twice and the eluates in 2 runs were collected and fractionated. The activity appeared around 27.1% acetonitrile for 1 ⁇ and around 27.6% acetonitrile for 2 ⁇ . Each of the active fractions was lyophilized and dissolved in 0.1 ml of DMSO.
  • EXAMPLE 8 Determination of amino acid sequences for the active substances showing the arachidonic acid metabolite release activity specific to the SENR-expression CHO cells purified from the swine spinal cord
  • Amino acid sequencing of the active substances showing the arachidonic acid metabolite release activity specific to the CHO/SENR cells purified in EXAMPLE 7 was performed. Since it was speculated that the active substances would be proteins or peptides as shown in EXAMPLE 6, amino-terminal amino acid sequencing was conducted by use of Procise 494 Protein Sequencer available from Perkin-Elmer, using the eluates containing the active peaks. As a result, the sequences shown by SEQ ID NO:5 and SEQ ID NO:6 were obtained. No amino acid was detected on the 6th and 11th residues.
  • the active substance 2 ⁇ was reduced and pyridylethylated using tributylphosphine and 4-vinylpyridine, which was then subjected to sequencing. No amino acid was yet detected on the 11th residue but pyridylethyl cysteine was detected on the 6th residue. From the foregoing it was surmised that the 6th and 11th residues of the active substance 2 ⁇ would be cysteine and these two Cys residues would form an intramolecular disulfide bond.
  • the active substance 1 ⁇ having a similar structure has cysteine on the 6th and 11th residues, speculating that these two Cys residues would likewise form an intramolecular disulfide bond.
  • SEQ ID NO:7 and SEQ ID NO:8 were deduced to represent the amino acid sequences of the two active substances.
  • SEQ ID NO:10 and SEQ ID NO:11 are partial sequences of the base sequence (accession No. AA535545) encoding a part of the precursor protein of human urotensin II (Coulouarn, Y. et al., Proc. Natl. Acad. Sci. USA, vol. 95, pp. 15803-15808 (1998)) registered in the GenBank database.
  • the reaction solution was composed of 1 ⁇ M each of the synthetic primers, 500 ng of the template DNA, 0.2 mM dNTPs, 1.25 unit of ExTaq DNA polymerase (Takara Shuzo Co.) and a buffer attached to the enzyme, which were mixed together to make the whole volume of the reaction solution 20 ⁇ l.
  • a cycle was set to include 94°C for 30 seconds, 60°C for 30 seconds and 72°C for 15 seconds, with 2 repetitions; a cycle of 94°C for 30 seconds, 55°C for 30 seconds and 72°C for 15 seconds, with 4 repetitions; a cycle of 94°C for 30 seconds, 52.5°C for 30 seconds and 72°C for 15 seconds, with 6 repetitions; and a cycle of 94°C for 30 seconds, 50°C for 30 seconds and 72°C for 15 seconds, with 30 repetitions.
  • the amplified product was confirmed by 1.5% agarose electrophoresis and ethidium bromide staining.
  • EXAMPLE 10 Determination of cDNA sequence containing the entire coding region of the SENR ligand precursor protein obtained from swine spinal cord cDNA library
  • RNAs were prepared from swine spinal cord. Then, poly(A) + RNA fraction was prepared using Oligotex(dT) 30 (Takara Shuzo Co.). From 2 ⁇ g of this poly(A) + RNA, cDNA was introduced into ⁇ Ziplox Not I/Sal I Arm, using Superscript Lamda System for cDNA Synthesis and ⁇ cloning kit (Gibco BRL), according to the manual attached. By packaging using Gigapack III Gold (Stratagene Co.), swine spinal cord cDNA library was prepared.
  • the filter was then incubated for 16 hours with the probes described below for hybridization.
  • the probes were selected from the sequence obtained in EXAMPLE 9, in which the forward strand was SEQ ID NO:13 and the reverse strand complementary in part to the former sequence was SEQ ID NO:14.
  • the synthesis of these probes were consigned (to Nippon Bio Service Co.). After denaturation of the probes at 70°C, the probes were slowly cooled to hybridize to one another, which was then radioactively labeled using Klenow enzyme in the presence of [ 32 P]dCTP (du Pont).
  • the labeled probe was further purified through a Nick column (Amersham Pharmacia Biotech Co.) and used for hybridization in a concentration of 1,000,000 cpm/ml.
  • the labeled probe was washed at room temperature 4 times with a 0.2 x SSC (diluted from 20 x SSC manufactured by Nippon Gene Co.) solution containing 0.1% SDS, and then twice at 65°C. Subsequently autoradiography was performed at -80°C to detect plaques hybridized. By this screening, hybridization signal was noted in 9 independent plaques.
  • the desired plasmid containing the swine SENR ligand precursor cDNA was excised from these positive plaques by the in vivo excision technique, according to the manual attached to Superscript Lamda System for cDNA Synthesis and ⁇ cloning kit (Gibco BRL) and transformed Escherichia coli XLlBlue.
  • the plasmid DNA was purified from the Escherichia coli.
  • the sequencing reaction for base sequence determination was carried out using DyeDeoxy Terminator Cycle Sequence Kit (Perkin-Elmer Co.).
  • the DNAs were decoded by an automated fluorescent sequencer.
  • three base sequences (SEQ ID NO:15, SEQ ID NO:16 and SEQ ID NO:17) encoding the entire sequence of the swine SENR ligand precursor protein were obtained.
  • a from the initiation codon ATG is made the first base, the 129th base is T in SEQ ID NO:15 and C in SEQ ID NO:16, but they were both translated into the same amino acid Asp (GAT, GAC).
  • SEQ ID NO:17 was found to be a splicing variant, in which C at the 101st to G at the 208th of SEQ ID NO:15 are deleted when the first base is A from the initiation codon ATG.
  • the corresponding amino acid sequences are: SEQ ID NO:18 for SEQ ID NO:15 and SEQ ID NO:16 and, SEQ ID NO:19 for SEQ ID NO:17. All of these precursor proteins were the precursors of swine SENR ligand 2 ⁇ (SEQ ID NO:8).
  • FIG. 8 shows the DNA sequence (SEQ ID NO:15) of swine SENR ligand precursor and the corresponding amino acid sequence (SEQ ID NO:18).
  • PCR was carried out using the primers shown by SEQ ID NO:10 and SEQ ID NO:11 employed in EXAMPLE 9.
  • the reaction solution was composed of 0.5 ⁇ M each of the synthetic primers, 500 ng of the template DNA, 0.2 mM dNTPs, 2.5 mM MgCl 2 , 0.2 ⁇ l of AmpliTaq Gold DNA polymerase (Perkin-Elmer Co.) and a buffer attached to the enzyme, which were mixed together to make the whole volume of the reaction solution 20 ⁇ l.
  • a cycle was set to include 94°C for 15 seconds, 60°C for 20 seconds and 72°C for 20 seconds, with 2 repetitions; a cycle of 94°C for 15 seconds, 55°C for 20 seconds and 72°C for 20 seconds, with 4 repetitions; a cycle of 94°C for 20 seconds, 52.5°C for 20 seconds and 72°C for 20 seconds, with 6 repetitions; a cycle of 94°C for 20 seconds, 50°C for 20 seconds and 72°C for 20 seconds, with 8 repetitions; and a cycle of 94°C for 30 seconds, 48°C for 20 seconds and 72°C for 20 seconds, with 30 repetitions, followed by maintaining at 72°C for 5 minutes.
  • the amplified product was confirmed by 1.5% agarose electrophoresis and ethidium bromide staining.
  • TOPO TA cloning kit Invitrogen Inc.
  • 2 ⁇ l of the reaction solution thus obtained was subcloned to plasmid vector pcr 2.1 and then introduced into Escherichia coli TOP10.
  • the plasmid DNA was purified using QIA prep8 mini prep (Qiagen Co.).
  • the reaction for base sequencing was carried out using DyeDeoxy Terminator Cycle Sequence Kit (Perkin-Elmer Co.). The DNAs were decoded using an automated fluorescent sequencer.
  • SEQ ID NO:20 which are considered to be a part of the swine SENR ligand precursor because of similarity to the sequence of swine SENR ligand precursor, was obtained as the PCR product.
  • the primer shown by SEQ ID NO:11 is a base sequence encoding a part of the ligand peptide and by comparing the amino acid sequence of swine SENR ligand, it was determined to be SEQ ID NO:21 as a bovine SENR ligand.
  • Boc-Val-OCH 2 -PAM resin (0.77 mmole/g resin) was charged in a reaction tank of peptide synthesizer ABI 430A. Thereafter, Boc-Cys(MeBzl), Boc-Tyr(Br-Z), Boc-Lys(Cl-Z), Boc-Trp(CHO), Boc-Phe, Boc-Cys(MeBzl), Boc-Glu(OcHex), Boc-Ser(Bzl), Boc-Thr(Bzl), Boc-Pro and Boc-Gly were introduced into the resin in this order according to the Boc-strategy (NMP-HOBt) peptide synthesis to give the desired protected peptide resin.
  • the resulting resin 0.59 g, was stirred at 0°C for 60 minutes in 10 ml of anhydrous hydrogen fluoride containing 2.22 g of p-cresol and 1.2 ml of 1,4-butanediol. Thereafter the hydrogen fluoride was distilled off in vacuum. Diethyl ether was added to the residue and the precipitate was filtrated. Aqueous 50% acetic acid solution was added to the precipitate for extraction and insoluble matters were removed. After the extract was sufficiently concentrated, the concentrate was applied to Sephadex G-25 (trade name) column (2.0 x 80 cm) filled with 50% acetic acid aqueous solution followed by development with the same solvent.
  • the main fractions were collected and applied to reverse phase chromatography column (2.6 x 60 cm) packed with LiChroprep (trade name) RP-18 followed by washing with 200 ml of water containing 0.1% TFA. Then linear density gradient elution was performed with 300 ml of 0.1% TFA and 300 ml of 40% acetonitrile containing 0.1% TFA. The main fractions were collected and concentrated. The concentrate was dissolved in approximately 4 ml of acetic acid. After diluting the solution with distilled water to a volume of 240 ml, pH was adjusted to 7.5 with ammonia water. The dilution was stirred while mildly blowing air therein. The reaction was traced by HPLC.
  • Human SENR ligand (SEQ ID NO:22) is the same peptide as reported to be human urotensin II (Coulouarn, Y. et al., Proc. Natl. Acad. Sci. USA, vol. 95, pp. 15803-15808 (1998)).
  • EXAMPLE 16 Arachidonic acid metabolite release-promoting activity of the synthetic swine SENR ligand polypeptides to the CHO/SENR cell line
  • the arachidonic acid metabolite release activity of the SENR ligand polypeptides 1 ⁇ and 2 ⁇ (SEQ ID NO:7 and SEQ ID NO:8) in various concentrations, which were synthesized in EXAMPLES 12 and 13, to the SENR-expression CHO cells was measured by the following method.
  • the CHO/SENR cells were inoculated on a 24-well plate in 5 x 10 4 cells/well. After incubation for 24 hours, [ 3 H] arachidonic acid was added to the system in 0.25 ⁇ Ci/well.
  • HBSS Hanks' balanced salt solution
  • BSA bovine serum albumin
  • the activity of the synthetic SENR ligand polypeptides 1 ⁇ and 2 ⁇ (SEQ ID NO:7 and SEQ ID NO:8) in various concentrations, which were synthesized in EXAMPLES 12 and 13, on blood pressure of rats under anesthesia was measured by the following method.
  • Male Wistar rats of 8 to 9 weeks old purchased from Nippon Crea Co.
  • Nembutal injection (Dai-Nippon Pharmaceutical Co., 50 mg/ml sodium pentobarbital, 50 mg/kg i.p.).
  • a catheter (SP-55) for blood pressure measurement connected with a transducer was inserted into the left carotid artery and a catheter (SP-35) for intravenous injection into the left femoral vein.
  • the synthetic ligand was dissolved in physiological saline containing 0.05% BSA, which was cannulated through the left femoral vein in a dose of 1, 10 or 100 nmol/kg.
  • the blood pressure was continuously recorded by means of a polygraph (manufactured by NEC Sanei Co.).
  • the blood pressure in rats decreased dose-dependently and the SENR ligand polypeptides showed a hypotensive action in rats.
  • Table 1 shows the hypotensive action on rat blood pressure (under anesthesia) when swine SENR ligand was administered in a dose of 10 nmols/kg.
  • the hypotensive action was shown by the average blood pressure reduced by administration of SENR ligand to the average blood pressure prior to administration.
  • EXAMPLE 18 Vasoconstrictive action of synthetic swine ligand polypeptide on rat thoracic aorta
  • the activity of the synthetic SENR ligand polypeptide 2 ⁇ (SEQ ID NO:8) in various concentrations, which were synthesized in EXAMPLE 13, on rat thoracic aorta was measured by the following method.
  • Male Wistar rats of 9 to 12 weeks old purchased from Nippon Charles River Co.
  • Nembutal injection (Dai-Nippon Pharmaceutical Co., 50 mg/ml sodium pentobarbital, 50 mg/kg i.p.).
  • the animal was bled to death by collecting whole blood from the abdominal aorta.
  • the thoracic aorta was removed from the rat to prepare ring preparations of 5 mm width.
  • the ring preparations were suspended in organ baths filled with 3 ml of Krebs-Henseleit solution (118 mM NaCl, 4.7 mM KCl, 2.5 mM CaCl 2 , 1.2 mM KH 2 PO 4 , 25 mM NaHCO 3 , 1.2 mM MgSO 4 , 10.0 mM glucose) kept at 37°C and gassed with a mixed gas (95%O 2 - 5%CO 2 ).
  • the isometric and contractive tension of each preparation was detected by a micro-load transducer (UL-10GR, Minebea) and recorded by a polygraph (NEC Sanei Co.). The resting tension was set to approximately 0.5 g.
  • the presence of endothelium was confirmed by observation that the constriction caused by administration of 10 -6 M norepinephrine was relaxed by administration of 10 -5 M acetylcholine.
  • the swine SENR ligand polypeptide was cumulatively administered to the final concentration of 10 -10 to 10 -7 M.
  • the rat thoracic aorta ring preparation was dose-dependently contracted by addition of the SENR ligand, as shown in FIG. 10.
  • a similar activity was confirmed as well, when using swine SENR ligand 1 ⁇ (SEQ ID NO:7), bovine SENR ligand (SEQ ID NO:21) or human SENR ligand (human urotensin II) (SEQ ID NO:22).
  • amplification by PCR was carried out using synthetic DNA primers shown by SEQ ID NO:23 and SEQ ID NO:24.
  • the synthetic DNA primers were so constructed as to amplify genes at the region to be translated into receptor proteins, whereupon recognition sites of the respective restriction enzymes were added at the 5' and 3' sides so that the base sequence recognized by restriction enzyme Sal I was added at the 5' side of the gene and the base sequence recognized by restriction enzyme Spe I at the 3' side.
  • the reaction solution was composed of 2.5 ⁇ l of the cDNA template, 0.2 ⁇ M each of the synthetic DNA primers, 0.2 mM dNTPs, 1 ⁇ l of Advantage2 polymerase mix (Clonetech Laboratories, Inc.) and a buffer attached to the enzyme, which were mixed together to make the total volume 50 ⁇ ml.
  • a cycle was set to include 95°C for 30 seconds and 72°C for 3 minutes, with 5 repetitions; then a cycle of 95°C for 30 seconds and 70°C for 3 minutes, with 5 repetitions; a further cycle of 95°C for 30 seconds and 68°C for 3 minutes, with 20 repetitions; and finally, heating was conducted at 94°C for 3 minutes.
  • the amplified products were confirmed by 0.8% agarose gel electrophoresis followed by ethidium bromide staining.
  • PCR products obtained after PCR in EXAMPLE 19 were separated by using a 0.8% low-melting temperature agarose gel. After the band parts were excised from the gel with a razor blade, DNAs were recovered using GENECLEAN SPIN (Bio 101 Co.). Following the protocol of Eukaryotic TOPOTM TA Cloning Kit (Invitrogen Co.), the recovered DNAs were subcloned into the plasmid vector, pcDNA3.1/V5/His to construct plasmid pcDNA3.1-hSENR for protein expression. The plasmid was introduced into Escherichia coli DH5 ⁇ competent cells (Toyobo Co.) to produce transformants.
  • clones carrying a cDNA-inserted fragment were selected in an LB agar medium containing ampicillin and picked up with a sterilized toothpick to obtain transformant E. coli DH5 ⁇ /pcDNA3.1-hSENR.
  • the individual clones were cultured overnight in an LB medium containing ampicillin.
  • Plasmid DNAs were prepared using Qiawell 8 Ultra Plasmid Kit (Qiagen Co.). An aliquot of the DNAs thus prepared was processed for cleavage by restriction enzyme Sal I to confirm the size and direction of the receptor inserted.
  • the reaction for base sequencing was carried out by using a DyeDeoxy Terminator Cycle Sequence Kit (Perkin-Elmer Co.), followed by decoding with a fluorescent automatic sequencer.
  • E. coli DH5 ⁇ /pcDNA3.1-hSENR prepared in EXAMPLE 20 was cultivated, plasmid DNA of pcDNA3.1-hSENR was prepared using Plasmid Midi Kit (Qiagen Co.). Using CellPhect Transfection Kit (Amersham Pharmacia Biotech Co.), the plasmid DNA was introduced into CHO dhfr - cells. Ten microgram of DNA was co-precipitated with calcium phosphate in suspension. The resulting suspension was added to a 10 cm Petri dish in which 5 ⁇ 10 5 or 1 ⁇ 10 6 CHO dhfr - cells had been seeded before 24 hours.
  • the cells were cultured for one day in MEM ⁇ containing 10% fetal calf serum. After passage, the cells were cultured in MEM ⁇ , as selection medium, containing 0.4 mg/ml of G418 (Gibco BRL) and 10% dialyzed fetal calf serum. Colonies of the transformant (CHO/hSENR) as human SENR-expression CHO cell, which grow in the selection medium, were selected.
  • the arachidonic acid metabolite release activity of the human SENR ligand polypeptide (human urotensin II) (SEQ ID NO:22) in various concentrations, which was synthesized in EXAMPLES 15, to the human SENR-expression CHO cells was measured by the following method. CHO/hSENR cells were inoculated on a 24-well plate in 5 x 10 4 cells/well. After incubation for 24 hours, [ 3 H] arachidonic acid was added to the system in 0.25 ⁇ Ci/well.
  • HBSS Hanks' balanced salt solution
  • BSA bovine serum albumin
  • the activity was expressed by percentage based on the amount of the arachidonic acid metabolites released in control group in which the buffer alone was added.
  • a similar activity was confirmed even when using swine SENR ligands (SEQ ID NO:7 and SEQ ID NO:8) and bovine SENR ligand (SEQ ID NO:21).
  • the [ 35 S] -guanosine 5'-( ⁇ -thio)triphosphate binding promoting activity of bovine SENR ligand (SEQ ID NO:21) on SENR-expression CHO cell membrane fraction was measured by the following method. First, preparation of membrane fraction is described. To 1 x 10 8 of CHO/SENR cells was added 10 ml of a homogenate buffer (10 mM NaHCO 3 , 5 mM EDTA, 0.5 mM PMSF, 1 ⁇ g/ml pepstatin, 4 ⁇ g/ml E64, 20 ⁇ g/ml leupeptin), which was homogenized by using Polytron (12,000 rpm, 1 min.).
  • a homogenate buffer (10 mM NaHCO 3 , 5 mM EDTA, 0.5 mM PMSF, 1 ⁇ g/ml pepstatin, 4 ⁇ g/ml E64, 20 ⁇ g/ml leupeptin
  • the cell homogenate was subjected to centrifugation (1,000 g, 15 mins.) to give the supernatant. Next, the supernatant was subjected to ultracentrifugation (Beckman type 30 rotor, 30,000 rpm, 1 hour). The resulting precipitate was used as a rat SENR-expression CHO cell membrane.
  • the GTP ⁇ S binding activity was measured as follows.
  • the rat SENR-expression CHO cell membrane was diluted with a buffer for membrane dilution (50 mM Tris-hydrochloride buffer (pH 7.4), 5 mM MgCl 2 , 150 mM NaCl, 1 ⁇ M GDP) to prepare a cell membrane fraction solution for assay having a protein concentration of 30 ⁇ g/ml.
  • a buffer for membrane dilution 50 mM Tris-hydrochloride buffer (pH 7.4), 5 mM MgCl 2 , 150 mM NaCl, 1 ⁇ M GDP
  • To 200 ⁇ l of the cell membrane fraction solution for assay were added 2 ⁇ l of 51.5 nM [ 35 S]-guanosine 5'-( ⁇ -thio)triphosphate (NEN Co.) and 2 ⁇ l of bovine SENR ligand (SEQ ID NO:21) in various concentrations.
  • the resulting solution mixture was kept at 25°C for an hour.
  • the mixture was filtrated through a filter.
  • a buffer for filter washing 50 mM Tris-hydrochloride buffer (pH 7.4), 5 mM MgCl2, 1 mM EDTA, 0.1% BSA
  • radioactivity of the filter was measured using a liquid scintillation counter.
  • the bovine SENR ligand dose-dependently increased the amount of [ 35 S]-guanosine 5'-( ⁇ -thio)triphosphate bound to the membrane fraction.
  • a similar activity was confirmed as well when using swine SENR ligands (SEQ ID NO: 7 and SEQ ID NO:8) or human SENR ligand (human urotensin II) (SEQ ID NO: 22).
  • the [ 35 S] -guanosine 5'-( ⁇ -thio)triphosphate binding promoting activity of human SENR ligand (human urotensin II) (SEQ ID NO:22) on human SENR-expression CHO cell membrane fraction was measured by the following method. First, preparation of membrane fraction is described. To 1 x 10 8 of CHO/hSENR cells was added 10 ml of a homogenate buffer (10 mM NaHCO 3 , 5 mM EDTA, 0.5 mM PMSF, 1 ⁇ g/ml pepstatin, 4 ⁇ g/ml E64, 20 ⁇ g/ml leupeptin), which was homogenized by using Polytron (12,000 rpm, 1 min.).
  • a homogenate buffer (10 mM NaHCO 3 , 5 mM EDTA, 0.5 mM PMSF, 1 ⁇ g/ml pepstatin, 4 ⁇ g/ml E64, 20 ⁇ g/ml leupeptin
  • the cell homogenate was subjected to centrifugation (1,000 g, 15 mins.) to give the supernatant. Next, the supernatant was subjected to ultracentrifugation (Beckman type 30 rotor, 30,000 rpm, an hour). The resulting precipitate was used as a human SENR-expression CHO cell membrane.
  • the GTP ⁇ S binding activity was measured as follows.
  • the human SENR-expression CHO cell membrane was diluted with a buffer for membrane dilution (50 mM Tris-hydrochloride buffer (pH 7.4), 5 mM MgCl2, 150 mM NaCl, 1 ⁇ M GDP) to prepare a cell membrane fraction solution for assay having a protein concentration of 30 ⁇ g/ml.
  • a buffer for membrane dilution 50 mM Tris-hydrochloride buffer (pH 7.4), 5 mM MgCl2, 150 mM NaCl, 1 ⁇ M GDP
  • To 200 ⁇ l of the cell membrane fraction solution for assay were added 2 ⁇ l of 51.5 nM [ 35 S]-guanosine 5'-( ⁇ -thio)triphosphate (NEN Co.) and 2 ⁇ l of human SENR ligand (SEQ ID NO:22) in various concentrations.
  • the resulting solution mixture was kept at 25°C for an hour.
  • the mixture was then filtrated through a filter.
  • a buffer for filter washing 50 mM Tris-hydrochloride buffer (pH 7.4), 5 mM MgCl 2 , 1 mM EDTA, 0.1% BSA
  • radioactivity of the filter was measured using a liquid scintillation counter.
  • the human SENR ligand dose-dependently increased the amount of [ 35 S] -guanosine 5'-( ⁇ -thio)triphosphate bound to the membrane fraction.
  • a similar activity was confirmed as well when using swine SENR ligands (SEQ ID NO:7 and SEQ ID NO:8) or bovine SENR ligand (SEQ ID NO:21).
  • Isotope-labeled bovine SENR ligand to use a binding inhibition test was prepared as follows. After 5 ⁇ g of bovine SENR ligand (SEQ ID NO:21) was dissolved in 25 ⁇ l of 0.4 M sodium acetate (pH 5.6), 200 ng of lacto-peroxidase (Wako Pure Chemicals Co.) was added to the solution and further 1 mCi [ 125 I] -sodium iodide (Amersham Pharmacia Biotech Co.) and 200 ng of hydrogen peroxide (10 ⁇ l) were added to the mixture. After allowing to stand at room temperature for 10 minutes, 200 ng of hydrogen peroxide (10 ⁇ l) were further added to the mixture followed by allowing to stand for 10 minutes.
  • reaction buffer A method for the binding inhibition test using [ 125 I]-labeled swine SENR ligands prepared in EXAMPLE 25 and rat SENR-expression CHO cells is shown below.
  • the CHO/SENR cells were inoculated on a 24-well plate in 5 ⁇ 10 4 cells/well followed by cultivation for 48 hours. The cells were then washed with 0.5 ml of MEM ⁇ medium containing 0.05% BSA (hereinafter the 0.05% BSA-containing MEM ⁇ medium is referred to as reaction buffer).
  • reaction buffer containing 200 pM [ 125 I]-labeled swine SENR ligand To the cells were added 0.5 ml each of the reaction buffer containing 200 pM [ 125 I]-labeled swine SENR ligand to examine the total binding, the reaction buffer containing 200 pM [ 125 I] -labeled swine SENR ligand and 1 ⁇ M of isotope-unlabeled bovine SENR ligand to examine non-specific binding and, the reaction buffer containing a test fluid and 200 pM [ 125 I]-labeled swine SENR ligand to examine the binding activity to SENR receptor, respectively, followed by reacting at room temperature for 30 minutes. After washing the cells with the reaction buffer, 0.2 ml of 0.5 N NaOH was added thereto to lyze the cells.
  • Radioactivity of the lyzed cells was measured with a gamma counter. Specific binding is obtained by reducing non-specific binding from the total binding.
  • the rat SENR receptor binding activity in the test fluid is shown by a ratio of binding obtained by reducing radioactivity of the test fluid-added lyzed cells from the total binding, to specific binding.
  • a method for the binding inhibition test using [ 125 I]-labeled human SENR ligand prepared by labeling human SENR ligand (SEQ ID NO:22) with [ 125 I] as in EXAMPLE 25 and human SENR-expression CHO cells is shown below.
  • CHO/hSENR cells were inoculated on a 24-well plate in 5 ⁇ 10 4 cells/well followed by cultivation for 48 hours. The cells were then washed with 0.5 ml of MEM ⁇ medium containing 0.05% BSA (hereinafter the 0.05% BSA-containing MEM ⁇ medium is referred to as reaction buffer).
  • reaction buffer containing 150 pM [ 125 I]-labeled human SENR ligand To the cells were added 0.5 ml each of the reaction buffer containing 150 pM [ 125 I]-labeled human SENR ligand to examine the total binding, the reaction buffer containing 150 pM [ 125 I]-labeled human SENR ligand and 1 ⁇ M of isotope-unlabeled human SENR ligand to examine non-specific binding and, the reaction buffer containing a test fluid and 150 pM [ 125 I]-labeled human SENR ligand to examine the binding activity to SENR receptor, respectively, followed by reacting at room temperature for 30 minutes. After washing the cells with the reaction buffer, 0.2 ml of 0.5 N NaOH was added thereto to lyze the cells.
  • Radioactivity of the lyzed cells was measured with a gamma counter. Specific binding is obtained by reducing non-specific binding from the total binding.
  • the human SENR receptor binding activity in the test fluid is shown by a ratio of binding obtained by reducing radioactivity of the test fluid-added lyzed cells from the total binding, to specific binding.
  • a method for the binding inhibition test using [ 125 I]-labeled bovine SENR ligand prepared in EXAMPLE 25 and rat SENR-expression CHO cell membrane fraction is shown below.
  • the membrane fraction prepared from CHO/SENR cells described in EXAMPLE 23 was diluted with a buffer for membrane dilution (50 mM Tris-hydrochloride buffer (pH 7.4), 5 mM MgCl 2 , 0.1% BSA, 5 mM EDTA, 0.5 mM PMSF, 1 ⁇ g/ml pepstatin, 4 ⁇ g/ml E64, 20 ⁇ g/ml leupeptin) to prepare the cell membrane fraction solution for assay having a protein concentration of 4 ⁇ g/ml.
  • a buffer for membrane dilution 50 mM Tris-hydrochloride buffer (pH 7.4), 5 mM MgCl 2 , 0.1% BSA, 5 mM EDTA, 0.5 mM PMSF, 1 ⁇ g
  • FIG. 12 shows the binding activity of bovine SENR ligand in various concentrations.
  • a method for the binding inhibition test using [ 125 I] -labeled human SENR ligand prepared in EXAMPLE 25 and human SENR-expression CHO cell membrane fraction is shown below.
  • the membrane fraction prepared from CHO/hSENR cells described in EXAMPLE 24 was diluted with a buffer for membrane dilution (50 mM Tris-hydrochloride buffer (pH 7.4), 5 mM MgCl 2 , 0.1% BSA, 5 mM EDTA, 0.5 mM PMSF, 1 ⁇ g/ml pepstatin, 4 ⁇ g/ml E64, 20 ⁇ g/ml leupeptin) to prepare the cell membrane fraction solution for assay having a protein concentration of 60 ⁇ g/ml.
  • a buffer for membrane dilution 50 mM Tris-hydrochloride buffer (pH 7.4), 5 mM MgCl 2 , 0.1% BSA, 5 mM EDTA, 0.5 mM PMSF, 1 ⁇ g
  • FIG. 13 shows the binding activity of human SENR ligand in various concentrations.
  • a method for the cAMP synthesis suppressing activity of the bovine SENR ligand (SEQ ID NO:21) synthesized in EXAMPLE 14 on rat SENR-expression CHO cells is shown below.
  • the CHO/SENR cells were inoculated on a 24-well plate in 5 ⁇ 10 4 cells/well followed by cultivation for 48 hours.
  • the cells were then washed with Hanks' buffer (pH 7.4) containing 0.2 mM 3-isobutyl-methylxanthine, 0.05% BSA and 20 mM HEPES (hereinafter Hanks' buffer (pH 7.4) containing 0.2 mM 3-isobutyl-methylxanthine, 0.05% BSA and 20 mM HEPES is referred to as a reaction buffer).
  • Hanks' buffer (pH 7.4) containing 0.2 mM 3-isobutyl-methylxanthine, 0.05% BSA and 20 mM HEPES is referred to as a reaction buffer).
  • 0.5 ml of the reaction buffer was added to the system, which was kept in the medium for 30 minutes.
  • the reaction buffer was removed and 0.25 ml of a fresh reaction buffer was added to the cells.
  • a method for the cAMP synthesis suppressing activity of the human SENR ligand (human urotensin II) (SEQ ID NO:22) synthesized in EXAMPLE 15 on human SENR-expression CHO cells is shown below.
  • the CHO/hSENR cells were inoculated on a 24-well plate in 5 ⁇ 10 4 cells/well followed by cultivation for 48 hours.
  • the cells were then washed with Hanks' buffer (pH 7.4) containing 0.2 mM 3-isobutyl-methylxanthine, 0.05% BSA and 20 mM HEPES (hereinafter Hanks' buffer (pH 7.4) containing 0.2 mM 3-isobutyl-methylxanthine, 0.05% BSA and 20 mM HEPES is referred to as a reaction buffer).
  • Hanks' buffer (pH 7.4) containing 0.2 mM 3-isobutyl-methylxanthine, 0.05% BSA and 20 mM HEPES is referred to as a reaction buffer).
  • 0.5 ml of the reaction buffer was added to the system, which was kept in the medium for 30 minutes.
  • the reaction buffer was removed and 0.25 ml of a fresh reaction buffer was added to the cells.
  • EXAMPLE 32 Determination of the partial sequence of swine SENR ligand precursor protein by PCR
  • PCR was carried out using the primers shown by SEQ ID NO:10 and SEQ ID NO:11.
  • the reaction solution was composed of 5 ⁇ M of the synthetic primer shown by SEQ ID NO:10, 1 ⁇ M of the synthetic primer shown by SEQ ID NO:11, 50 ng of the template DNA, 0.2 mM dNTPs, 2.5 mM MgCl 2 , 0.4 ⁇ l of AmpliTaq Gold DNA polymerase (Perkin-Elmer Co.) and 1/10 volume of 10-fold concentrated AmpliTaq Gold buffer, which were mixed together to make the whole volume of the reaction solution 40 ⁇ l.
  • a cycle was set to include 94°C for 15 seconds, 60°C for 20 seconds and 72°C for 20 seconds, with 4 repetitions; a cycle of 94°C for 15 seconds, 52.5°C for 20 seconds and 72°C for 20 seconds, with 6 repetitions; a cycle of 94°C for 20 seconds, 52.5°C for 20 seconds and 72°C for 20 seconds, with 6 repetitions; a cycle of 94°C for 20 seconds, 50°C for 20 seconds and 72°C for 20 seconds, with 8 repetitions; and a cycle of 94°C for 15 seconds, 48°C for 20 seconds and 72°C for 20 seconds, with 30 repetitions, followed by maintaining at 72°C for 7 minutes.
  • EXAMPLE 34 Acquisition of the 5' sequence of the gene encoding bovine SENR ligand precursor protein by 5' RACE method
  • PCR was carried out using adapter primer AP1 attached to Marathon cDNA amplification kit (Clonetech Laboratories, Inc.) and the primer shown by SEQ ID NO:31.
  • the reaction solution was composed of 0.5 ⁇ M of the primer shown by SEQ ID NO:31, 0.2 ⁇ M of AP1, 0.2 mM dNTPs, 2.5 mM MgCl 2 , 0.2 ⁇ l of AmpliTaq Gold DNA polymerase (Perkin-Elmer Co.) and 1/10 volume of 10-fold concentrated AmpliTaq Gold buffer, which were mixed together to make the whole volume of the reaction solution 20 ⁇ l.
  • Using Thermal Cycler (Perkin-Elmer Co.) for amplification after maintaining at 95°C for 9 minutes, a cycle was set to include 95°C for 10 seconds and 68°C for 1 minute, with 40 repetitions.
  • reaction solution was composed of 0.5 ⁇ M of the primer shown by SEQ ID NO:32, 0.2 ⁇ M of AP2, 0.2 mM dNTPs, 2.5 mM MgCl 2 , 0.2 ⁇ l of AmpliTaq Gold DNA polymerase (Perkin-Elmer Co.) and 1/10 volume of 10-fold concentrated AmpliTaq Gold buffer, which were mixed together to make the whole volume of the reaction solution 20 ⁇ l.
  • a cycle was set to include 95°C for 10 seconds and 66°C for 1 minute, with 40 repetitions, followed by maintaining at 72°C for 10 minutes.
  • the PCR solution was subjected to electrophoresis using 3.5% Nusieve GTG Agarose (Takara Shuzo Co.) and DNA was extracted from the bands around 420 bp, which were detected by ethidium bromide staining, using GeneClean Spin kit (Bio 101 Co.).
  • the DNA was subcloned to plasmid pcr 2.1 using TOPO TA cloning kit (Invitrogen Co.) and then introduced into Escherichia coli TOPO10. From the resulting transformant, the plasmid DNA was purified using QIA prep8 mini prep kit (Qiagen Co.). The reaction for base sequencing was carried out using DyeDeoxy Terminator Cycle Sequence Kit (Perkin-Elmer Co.). Decoding by an automated fluorescent sequencer gave the sequence shown by SEQ ID NO: 33.
  • PCR was carried out using adapter primer AP1 attached to Marathon cDNA amplification kit (Clonetech Laboratories, Inc.) and the primer shown by SEQ ID NO:34.
  • the reaction solution was composed of 0.2 ⁇ M of the primer shown by SEQ ID NO:34, 0.2 ⁇ M of AP1, 0.2 mM dNTPs, 2.5 mM MgCl 2 , 0.2 ⁇ l of AmpliTaq Gold DNA polymerase (Perkin-Elmer Co.) and 1/10 volume of 10-fold concentrated AmpliTaq Gold buffer, which were mixed together to make the whole volume of the reaction solution 20 ⁇ l.
  • Using Thermal Cycler (Perkin-Elmer Co.) for amplification after maintaining at 95°C for 9 minutes, a cycle was set to include 95°C for 10 seconds and 68°C for 1 minute, with 40 repetitions.
  • reaction solution was composed of 0.2 ⁇ M of the primer shown by SEQ ID NO:35, 0.2 ⁇ M of AP2, 0.2 mM dNTPs, 2.5 mM MgCl 2 , 0.2 ⁇ l of AmpliTaq Gold DNA polymerase (Perkin-Elmer Co.) and 1/10 volume of 10-fold concentrated AmpliTaq Gold buffer, which were mixed together to make the whole volume of the reaction solution 20 ⁇ l.
  • a cycle was set to include 95°C for 10 seconds and 66°C for 1 minute, with 40 repetitions, followed by maintaining at 72°C for 10 minutes.
  • the PCR solution was subjected to electrophoresis using 3.5% Nusieve GTG Agarose (Takara Shuzo Co.) and DNA was extracted from the bands around 300 bp, which were detected by ethidium bromide staining, using GeneClean Spin kit (Bio 101 Co.).
  • the DNA was subcloned to plasmid pcr 2.1 using TOPO TA cloning kit (Invitrogen Co.) and then introduced into Escherichia coli TOPO10. From the resulting transformant, the plasmid DNA was purified using QIA prep8 mini prep kit (Qiagen Co.). The reaction for base sequencing was carried out using DyeDeoxy Terminator Cycle Sequence Kit (Perkin-Elmer Co.). Decoding by an automated fluorescent sequencer gave the sequence shown by SEQ ID NO:36.
  • PCR was carried out using the primers shown by SEQ ID NO:37 and SEQ ID NO:38, in which an aliquot of the double-stranded cDNA solution corresponding to 4 ng of mRNA was used as a template.
  • the reaction solution was composed of 0.5 ⁇ M each of the two primers, 0.2 mM dNTPs, 2.5 mM MgCl 2 , 0.2 ⁇ l of AmpliTaq Gold DNA polymerase (Perkin-Elmer Co.) and 1/10 volume of 10-fold concentrated AmpliTaq Gold buffer, which were mixed together to make the whole volume of the reaction solution 20 ⁇ l.
  • Using Thermal Cycler (Perkin-Elmer Co.) for amplification after maintaining at 95°C for 9 minutes, a cycle was set to include 95°C for 10 seconds, 62°C for 20 seconds and 72°C for 1 minute, with 40 repetitions, followed by maintaining at 72°C for 10 minutes.
  • the PCR solution was subjected to electrophoresis using 3.5% Nusieve GTG Agarose (Takara Shuzo Co.) and DNA was extracted from the bands around 490 bp, which were detected by ethidium bromide staining, using GeneClean Spin kit (Bio 101 Co.).
  • the DNA was subcloned to plasmid pcr 2.1 using TOPO TA cloning kit (Invitrogen Co.) and then introduced into Escherichia coli TOPO10. From the resulting transformant, the plasmid DNA was purified using QIA prep8 mini prep kit (Qiagen Co.).
  • the reaction for base sequencing was carried out using DyeDeoxy Terminator Cycle Sequence Kit (Perkin-Elmer Co.).
  • SEQ ID NO:30 Decoding by an automated fluorescent sequencer gave the sequence shown by SEQ ID NO:30. In this sequence, the full length of bovine SENR ligand precursor was contained. Thus, Escherichia coli TOP10 was transformed by this plasmid to obtain Escherichia coli TOP10/pCR-buro.
  • the amino acid sequence of the bovine SENR ligand precursor protein translated from SEQ ID NO:30 are shown in SEQ ID NO:29.
  • amino acid sequence of bovine SENR ligand deduced from the amino acid sequence of bovine SENR ligand precursor protein is shown by SEQ ID NO:21 and the base sequence encoding the same is shown by SEQ ID NO:28, respectively.
  • the total base sequence and amino acid sequence of bovine SENR ligand precursor protein are shown in FIG. 14.
  • urotensin II SEQ ID NO:39
  • Cys-Phe-Trp-Lys-Tyr-Cys-Val urotensin II
  • bovine and human SENR ligand polypeptides an antibody capable of recognizing the C terminus of SENR ligand polypeptides was prepared.
  • “Haze” urotensin II peptide, 1 mg, and 4 mg of BTG (bovine thyroglobulin) were bound to one another, using 30 mg of ECDI (1-ethyl-3-(3-dimethylaminopropyl) -carbodiimide, Dojin Kagaku Co.) to produce the "Haze” urotensin II-carrier protein complex.
  • the "Haze” urotensin II-carrier protein complex was dialyzed against 0.15 M NaCl aqueous solution, which was then mixed with dialysate and Freund's complete adjuvant.
  • mice Female, 6 to 8 weeks old
  • mice were primary immunized with "Haze" urotensin II in 20 ⁇ g/mouse.
  • the complex was mixed with Freund's incomplete adjuvant, which was used as an antigen for second immunization.
  • the animal was immunized with a mixture of the complex and Freund's incomplete adjuvant every 2 weeks until its antibody titer increased.
  • the antibody titer was assayed by enzyme immunoassay utilizing biotinylated "Haze” urotensin II peptide.
  • the biotinylated “Haze” urotensin II peptide [N-biotinyl-Ala 1 ]-urotensin II was obtained by subjecting the reaction product of NHS-biotin (N-hydroxysuccinimidobiotin) and "Haze” urotensin II peptide to preparative HPLC.
  • the well was washed and a peroxidase substrate was added to the well, whereby a color generated on the substrate was measured with a 96-well multi-photometer.
  • the sera added to the wells in which the substrate generated a color was determined to be sera with an increased antibody titer. Since the antibody detected here binds to the N-terminal labeled biotinylated "Haze" urotensin II, the antibody is considered to recognize the C-terminal structure of the peptide.
  • the antibody contained in these sera is capable of recognizing swine, bovine and human SENR ligand polypeptides.
  • the DNA encoding the polypeptide of the present invention or the polypeptide of the present invention can be used: (1) for the survey of physiological activities that the polypeptide of the present invention possesses, (2) for preparing synthetic oligonucleotide probes or primers for PCR, (3) for acquiring DNAs encoding ligands to SENR or precursor proteins, (4) for the development of the receptor-binding assay system using the expression system of recombinant receptor protein and screening of a candidate drug, (5) for acquiring antibodies and antisera, (6) for the development of diagnostic agents using DNAs, RNAs, antibodies or antisera, (7) for the development of drugs such as agents for regulating central nervous functions, circulatory functions, heart functions, renal functions, urinary functions, sensory functions, etc., (8) for gene therapy, and the like.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Organic Chemistry (AREA)
  • Medicinal Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Engineering & Computer Science (AREA)
  • Animal Behavior & Ethology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Neurosurgery (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Biomedical Technology (AREA)
  • Neurology (AREA)
  • Heart & Thoracic Surgery (AREA)
  • Urology & Nephrology (AREA)
  • Endocrinology (AREA)
  • Toxicology (AREA)
  • Zoology (AREA)
  • Cardiology (AREA)
  • Biochemistry (AREA)
  • Biophysics (AREA)
  • Genetics & Genomics (AREA)
  • Molecular Biology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Peptides Or Proteins (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
EP99973037A 1998-11-30 1999-11-29 Neuartige, physiologisch wirksame substanz, verfahren zu deren herstellung und anwendung derselben Withdrawn EP1136503A1 (de)

Applications Claiming Priority (7)

Application Number Priority Date Filing Date Title
JP33898498 1998-11-30
JP33898498 1998-11-30
JP2684899 1999-02-04
JP2684899 1999-02-04
JP23936799 1999-08-26
JP23936799 1999-08-26
PCT/JP1999/006649 WO2000032627A1 (fr) 1998-11-30 1999-11-29 Nouvelle substance physiologiquement active et ses procedes d'obtention et d'utilisation

Publications (1)

Publication Number Publication Date
EP1136503A1 true EP1136503A1 (de) 2001-09-26

Family

ID=27285566

Family Applications (1)

Application Number Title Priority Date Filing Date
EP99973037A Withdrawn EP1136503A1 (de) 1998-11-30 1999-11-29 Neuartige, physiologisch wirksame substanz, verfahren zu deren herstellung und anwendung derselben

Country Status (4)

Country Link
EP (1) EP1136503A1 (de)
AU (1) AU1411200A (de)
CA (1) CA2352760A1 (de)
WO (1) WO2000032627A1 (de)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2004037863A1 (ja) * 2002-10-25 2004-05-06 Takeda Pharmaceutical Company Limited 抗体およびその用途

Families Citing this family (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
AU5848400A (en) * 1999-07-08 2001-01-30 Takeda Chemical Industries Ltd. Novel physiologically active substance, process for producing the same and use thereof
EP1262195A4 (de) * 2000-03-07 2003-05-21 Takeda Chemical Industries Ltd Vasoaktieve agentia
AU2001271018A1 (en) * 2000-07-04 2002-01-14 Takeda Chemical Industries Ltd. Gpr14 antagonist
EP1308513A4 (de) * 2000-08-10 2008-07-09 Takeda Pharmaceutical Anwendung von polypeptiden
EP1331010A4 (de) * 2000-08-25 2008-08-06 Takeda Pharmaceutical Mittel zur prävention und behandlung von erkrankungen des zentralen nervensystems

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1999035266A2 (en) * 1998-01-09 1999-07-15 Smithkline Beecham Corporation Human urotensin ii

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
See references of WO0032627A1 *

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2004037863A1 (ja) * 2002-10-25 2004-05-06 Takeda Pharmaceutical Company Limited 抗体およびその用途

Also Published As

Publication number Publication date
AU1411200A (en) 2000-06-19
WO2000032627A1 (fr) 2000-06-08
CA2352760A1 (en) 2000-06-08

Similar Documents

Publication Publication Date Title
US7799533B2 (en) Polypeptides, their production and use
US7608409B2 (en) Screening assay using G-protein coupled receptor protein OT7T175
US7419956B2 (en) Isolated physiologically active peptide and use thereof
US7374910B2 (en) DNA encoding galanin receptor activating peptide
CA2387711A1 (en) Novel g protein-coupled receptor protein and dna thereof
EP1132405A1 (de) Neuartiges g-protein gekoppeltes rezeptorprotein, dessen dna und ligand
EP1293567B1 (de) Gpr8 ligand und dafuer kodierende dna
EP1179347A1 (de) Kontrollsubstanzen für die leberfunktion
EP1136503A1 (de) Neuartige, physiologisch wirksame substanz, verfahren zu deren herstellung und anwendung derselben
US7662574B2 (en) Use of a G protein-coupled receptor and its cognizant ligand in the identification of compounds that affect prolactin secretion
EP1394543B1 (de) Screening-verfahren
EP1559721B1 (de) Fprl1-liganden und deren verwendung
JP4429410B2 (ja) 新規生理活性物質、その製造法および用途
WO2001004298A1 (fr) Nouvelle substance physiologiquement active, procede de production et utilisation
JP2001128688A (ja) ポリペプチド、その製造法および用途
EP1227105A1 (de) Ghsr-polypeptidliganden und für diese kodierende dna
JP2001069996A (ja) 新規生理活性物質、その製造法および用途
JP2000157273A (ja) 新規生理活性ペプチドおよびその用途
ZA200103152B (en) G Protein-coupled receptor proteins, DNAS thereof and ligands to the same.
MXPA01004177A (en) Novel g protein-coupled receptor proteins, dnas thereof and ligands to the same

Legal Events

Date Code Title Description
PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

17P Request for examination filed

Effective date: 20010523

AK Designated contracting states

Kind code of ref document: A1

Designated state(s): AT BE CH CY DE DK ES FI FR GB GR IE IT LI LU MC NL PT SE

AX Request for extension of the european patent

Free format text: AL;LT;LV;MK;RO;SI

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE APPLICATION HAS BEEN WITHDRAWN

18W Application withdrawn

Effective date: 20040211